1
|
Malekzadeh H, Tirmizi Z, Arellano JA, Egro FM, Ejaz A. Application of Adipose-Tissue Derived Products for Burn Wound Healing. Pharmaceuticals (Basel) 2023; 16:1302. [PMID: 37765109 PMCID: PMC10534650 DOI: 10.3390/ph16091302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Burn injuries are a significant global health concern, leading to high morbidity and mortality. Deep burn injuries often result in delayed healing and scar formation, necessitating effective treatment options. Regenerative medicine, particularly cell therapy using adipose-derived stem cells (ASCs), has emerged as a promising approach to improving burn wound healing and reducing scarring. Both in vitro and preclinical studies have demonstrated the efficacy of ASCs and the stromal vascular fraction (SVF) in addressing burn wounds. The application of ASCs for burn healing has been studied in various forms, including autologous or allogeneic cells delivered in suspension or within scaffolds in animal burn models. Additionally, ASC-derived non-cellular components, such as conditioned media or exosomes have shown promise. Injection of ASCs and SVF at burn sites have been demonstrated to enhance wound healing by reducing inflammation and promoting angiogenesis, epithelialization, and granulation tissue formation through their paracrine secretome. This review discusses the applications of adipose tissue derivatives in burn injury treatment, encompassing ASC transplantation, as well as the utilization of non-cellular components utilization for therapeutic benefits. The application of ASCs in burn healing in the future will require addressing donor variability, safety, and efficacy for successful clinical application.
Collapse
Affiliation(s)
| | | | | | | | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
2
|
Saranya I, Akshaya R, Selvamurugan N. Regulation of Wnt signaling by non-coding RNAs during osteoblast differentiation. Differentiation 2022; 128:57-66. [DOI: 10.1016/j.diff.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
|
3
|
Wang P, Deng Z, Li A, Li R, Huang W, Cui J, Chen S, Li B, Zhang S. β-Catenin promotes long-term survival and angiogenesis of peripheral blood mesenchymal stem cells via the Oct4 signaling pathway. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1434-1449. [PMID: 36050404 PMCID: PMC9535028 DOI: 10.1038/s12276-022-00839-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 04/06/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022]
Abstract
Stem cell therapy has been extensively studied to improve heart function following myocardial infarction; however, its therapeutic potency is limited by low rates of engraftment, survival, and differentiation. Here, we aimed to determine the roles of the β-catenin/Oct4 signaling axis in the regulation of long-term survival and angiogenesis of peripheral blood mesenchymal stem cells (PBMSCs). These cells were obtained from rat abdominal aortic blood. We showed that β-catenin promotes the self-renewal, antiapoptotic effects, and long-term survival of PBMSCs by activating the Oct4 pathway through upregulation of the expression of the antiapoptotic factors Bcl2 and survivin and the proangiogenic cytokine bFGF and suppression of the levels of the proapoptotic factors Bax and cleaved caspase-3. β-Catenin overexpression increased Oct4 expression. β-Catenin knockdown suppressed Oct4 expression in PBMSCs. However, β-catenin levels were not affected by Oct4 overexpression or knockdown. Chromatin immunoprecipitation assays proved that β-catenin directly regulates Oct4 transcription in PBMSCs. In vivo, PBMSCs overexpressing β-catenin showed high survival in infarcted hearts and resulted in better myocardial repair. Further functional analysis identified Oct4 as the direct upstream regulator of Ang1, bFGF, HGF, VEGF, Bcl2, and survivin, which cooperatively drive antiapoptosis and angiogenesis of engrafted PBMSCs. These findings revealed the regulation of β-catenin in PBMSCs by the Oct4-mediated antiapoptotic/proangiogenic signaling axis and provide a breakthrough point for improving the long-term survival and therapeutic effects of PBMSCs. Boosting expression of a specific gene has allowed researchers to generate stem cells with increased capacity for tissue repair after a heart attack. Several studies have shown that treatment with a population of circulating cells known as ‘peripheral blood mesenchymal stem cells’ (PBMSCs) can regenerate cardiac tissue. These cells generally have a short lifespan when used therapeutically, but researchers led by Shaoheng Zhang at Jinan University in Guangzhou China have increased long-term survival and performance by boosting expression of the gene encoding β-catenin, a protein that promotes cell survival and proliferation. PBMSCs expressing increased levels of β-catenin preserved heart function in a rat model of heart attack, stimulating blood vessel growth and improving animal survival. This study also reveals proteins regulated by β-catenin, which could potentially be exploited for finer control of PBMSC function.
Collapse
Affiliation(s)
- Pengzhen Wang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China.,Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Zhanyu Deng
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Aiguo Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Rongsen Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Weiguang Huang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Jin Cui
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Songsheng Chen
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Biao Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China
| | - Shaoheng Zhang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, Guangdong, 510220, P.R. China.
| |
Collapse
|
4
|
Abdul Kareem N, Aijaz A, Jeschke MG. Stem Cell Therapy for Burns: Story so Far. Biologics 2021; 15:379-397. [PMID: 34511880 PMCID: PMC8418374 DOI: 10.2147/btt.s259124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Burn injuries affect approximately 11 million people annually, with fatalities amounting up to 180,000. Burn injuries constitute a global health issue associated with high morbidity and mortality. Recent years have seen advancements in regenerative medicine for burn wound healing encompassing stem cells and stem cell-derived products such as exosomes and conditioned media with promising results compared to current treatment approaches. Sources of stem cells used for treatment vary ranging from hair follicle stem cells, embryonic stem cells, umbilical cord stem cells, to mesenchymal stem cells, such as adipose-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, and even stem cells harvested from discarded burn tissue. Stem cells utilize various pathways for wound healing, such as PI3/AKT pathway, WNT-β catenin pathway, TGF-β pathway, Notch and Hedgehog signaling pathway. Due to the paracrine signaling mechanism of stem cells, exosomes and conditioned media derived from stem cells have also been utilized in burn wound therapy. As exosomes and conditioned media are cell-free therapy and contain various biomolecules that facilitate wound healing, they are gaining popularity as an alternative treatment strategy with significant improvement in outcomes. The treatment is provided either as direct injections or embedded in a natural/artificial scaffold. This paper reviews in detail the different sources of stem cells, stem cell-derived products, their efficacy in burn wound repair, associated signaling pathways and modes of delivery for wound healing.
Collapse
Affiliation(s)
| | - Ayesha Aijaz
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Marc G Jeschke
- Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
5
|
Yin S, Cao Y. Hydrogels for Large-Scale Expansion of Stem Cells. Acta Biomater 2021; 128:1-20. [PMID: 33746032 DOI: 10.1016/j.actbio.2021.03.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summarize current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.
Collapse
Affiliation(s)
- Sheng Yin
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057.
| |
Collapse
|
6
|
de Winter TJJ, Nusse R. Running Against the Wnt: How Wnt/β-Catenin Suppresses Adipogenesis. Front Cell Dev Biol 2021; 9:627429. [PMID: 33634128 PMCID: PMC7900430 DOI: 10.3389/fcell.2021.627429] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) give rise to adipocytes, osteocytes, and chondrocytes and reside in various tissues, including bone marrow and adipose tissue. The differentiation choices of MSCs are controlled by several signaling pathways, including the Wnt/β-catenin signaling. When MSCs undergo adipogenesis, they first differentiate into preadipocytes, a proliferative adipocyte precursor cell, after which they undergo terminal differentiation into mature adipocytes. These two steps are controlled by the Wnt/β-catenin pathway, in such a way that when signaling is abrogated, the next step in adipocyte differentiation can start. This sequence suggests that the main role of Wnt/β-catenin signaling is to suppress differentiation while increasing MSC and preadipocytes cell mass. During later steps of MSC differentiation, however, active Wnt signaling can promote osteogenesis instead of keeping the MSCs undifferentiated and proliferative. The exact mechanisms behind the various functions of Wnt signaling remain elusive, although recent research has revealed that during lineage commitment of MSCs into preadipocytes, Wnt signaling is inactivated by endogenous Wnt inhibitors. In part, this process is regulated by histone-modifying enzymes, which can lead to increased or decreased Wnt gene expression. The role of Wnt in adipogenesis, as well as in osteogenesis, has implications for metabolic diseases since Wnt signaling may serve as a therapeutic target.
Collapse
Affiliation(s)
- Twan J J de Winter
- Faculty of Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Roeland Nusse
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford, CA, United States.,School of Medicine, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
7
|
Pectin-based self-healing hydrogel with NaHCO3 degradability for drug loading and release. JOURNAL OF POLYMER RESEARCH 2021. [DOI: 10.1007/s10965-021-02430-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
8
|
Sutjarit N, Thongon N, Weerachayaphorn J, Piyachaturawat P, Suksamrarn A, Suksen K, Papachristou DJ, Blair HC. Inhibition of Adipogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells by a Phytoestrogen Diarylheptanoid from Curcuma comosa. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:9993-10002. [PMID: 32838526 DOI: 10.1021/acs.jafc.0c04063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We investigated the effect of a phytoestrogen, (3R)-1,7-diphenyl-(4E,6E)-4,6-heptadien-3-ol (DPHD), from Curcuma comosa Roxb. (Zingiberaceae family) on the adipogenic differentiation of mesenchymal progenitors, human bone marrow-derived mesenchymal stem cells (hBMSCs). DPHD inhibited adipocyte differentiation of hBMSCs by suppressing the expression of genes involved in adipogenesis. DPHD at concentrations of 0.1, 1, and 10 μM significantly decreased triglyceride accumulation in hBMSCs to 7.1 ± 0.2, 6.3 ± 0.4, and 4.9 ± 0.2 mg/dL, respectively, compared to the nontreated control (10.1 ± 0.9 mg/dL) (p < 0.01). Based on gene expression profiling, DPHD increased the expression of several genes involved in the Wnt/β-catenin signaling pathway, a negative regulator of adipocyte differentiation in hBMSCs. DPHD also increased the levels of essential signaling proteins which are extracellular signal-regulated kinases 1 and 2 (ERK1/2) and glycogen synthase kinase 3 beta (GSK-3β) that link estrogen receptor (ER) signaling to Wnt/β-catenin signaling. In conclusion, DPHD exhibited the anti-adipogenic effect in hBMSCs by suppression of adipogenic markers in hBMSCs through the activation of ER and Wnt/β catenin signaling pathways. This finding suggests the potential role of DPHD in preventing bone marrow adiposity which is one of the major factors that exacerbates osteoporosis in postmenopause.
Collapse
Affiliation(s)
- Nareerat Sutjarit
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Natthakan Thongon
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Pawinee Piyachaturawat
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Kanoknetr Suksen
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Dionysios J Papachristou
- Laboratory of Bone and Soft Tissue Studies, Department of Anatomy-Histology-Embryology, University Patras Medical School, Patras 26504, Greece
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Harry C Blair
- The Pittsburgh Veterans Affairs Medical Center, Pittsburgh, Pennsylvania 15261, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
9
|
Schupbach D, Comeau-Gauthier M, Harvey E, Merle G. Wnt modulation in bone healing. Bone 2020; 138:115491. [PMID: 32569871 DOI: 10.1016/j.bone.2020.115491] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Genetic studies have been instrumental in the field of orthopaedics for finding tools to improve the standard management of fractures and delayed unions. The Wnt signaling pathway that is crucial for development and maintenance of many organs also has a very promising pathway for enhancement of bone regeneration. The Wnt pathway has been shown to have a direct effect on stem cells during bone regeneration, making Wnt a potential target to stimulate bone repair after trauma. A more complete view of how Wnt influences animal bone regeneration has slowly come to light. This review article provides an overview of studies done investigating the modulation of the canonical Wnt pathway in animal bone regeneration models. This not only includes a summary of the recent work done elucidating the roles of Wnt and β-catenin in fracture healing, but also the results of thirty transgenic studies, and thirty-eight pharmacological studies. Finally, we discuss the discontinuation of sclerostin clinical trials, ongoing clinical trials with lithium, the results of Dkk antibody clinical trials, the shift into combination therapies and the future opportunities to enhance bone repair and regeneration through the modulation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Drew Schupbach
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Marianne Comeau-Gauthier
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Edward Harvey
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada.
| | - Geraldine Merle
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Department of Chemical Engineering, Polytechnique Montreal, 2500, chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada.
| |
Collapse
|
10
|
Chen J, Ashames A, Buabeid MA, Fahelelbom KM, Ijaz M, Murtaza G. Nanocomposites drug delivery systems for the healing of bone fractures. Int J Pharm 2020; 585:119477. [PMID: 32473968 DOI: 10.1016/j.ijpharm.2020.119477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/20/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
The skeletal system is fundamental for the structure and support of the body consisting of bones, cartilage, and connective tissues. Poor fracture healing is a chief clinical problem leading to disability, extended hospital stays and huge financial liability. Even though most fractures are cured using standard clinical methods, about 10% of fractures are delayed or non-union. Despite decades of progress, the bone-targeted delivery system is still restricted due to the distinctive anatomical bone features. Recently, various novel nanocomposite systems have been designed for the cell-specific targeting of bone, enhancing drug solubility, improving drug stability and inhibiting drug degradation so that it can reach its target site without being removed in the systemic circulation. Such targeting systems could consist of biological compounds i.e. bone marrow stem cells (BMSc), growth factors, RNAi, parathyroid hormone or synthetic compounds, i.e. bisphosphonates (BPs) and calcium phosphate cement. Hydrogels and nanoparticles are also being employed for fracture healing. In this review, we discussed the normal mechanism of bone healing and all the possible drug delivery systems being employed for the healing of the bone fracture.
Collapse
Affiliation(s)
- Jianxian Chen
- School of Economics, Capital University of Economics and Business, Beijing, China
| | - Akram Ashames
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates.
| | - Manal Ali Buabeid
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Khairi Mustafa Fahelelbom
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - Muhammad Ijaz
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan.
| |
Collapse
|
11
|
Haugen HJ, Lyngstadaas SP, Rossi F, Perale G. Bone grafts: which is the ideal biomaterial? J Clin Periodontol 2019; 46 Suppl 21:92-102. [DOI: 10.1111/jcpe.13058] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Håvard Jostein Haugen
- Department of Biomaterials; Institute of Clinical Dentistry; Faculty of Dentistry; University of Oslo; Oslo Norway
| | - Ståle Petter Lyngstadaas
- Department of Biomaterials; Institute of Clinical Dentistry; Faculty of Dentistry; University of Oslo; Oslo Norway
- Corticalis AS; Oslo Science Park Oslo Norway
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Milano Italy
| | - Giuseppe Perale
- Industrie Biomediche Insubri SA; Mezzovico-Vira Switzerland
- Biomaterials Laboratory; Institute for Mechanical Engineering and Materials Technology; University of Applied Sciences and Arts of Southern Switzerland; Manno Switzerland
- Department of Surgical Sciences; Faculty of Medical Sciences; Orthopaedic Clinic-IRCCS A.O.U. San Martino; Genova Italy
| |
Collapse
|
12
|
Unveiling Mesenchymal Stromal Cells' Organizing Function in Regeneration. Int J Mol Sci 2019; 20:ijms20040823. [PMID: 30769851 PMCID: PMC6413004 DOI: 10.3390/ijms20040823] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/03/2019] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
Regeneration is a fundamental process attributed to the functions of adult stem cells. In the last decades, delivery of suspended adult stem cells is widely adopted in regenerative medicine as a leading means of cell therapy. However, adult stem cells cannot complete the task of human body regeneration effectively by themselves as far as they need a receptive microenvironment (the niche) to engraft and perform properly. Understanding the mechanisms underlying mammalian regeneration leads us to an assumption that improved outcomes of cell therapy require a specific microenvironment that is generated in damaged areas prior to stem cell delivery. To a certain extent, it may be achieved by the delivery of mesenchymal stromal cells (MSCs), not in dispersed form, but rather in self-organized cell sheets (CS) ⁻ tissue-like structures comprised of viable cells and microenvironment components: extracellular matrix and soluble factors deposited in the matrix. In this review, we highlight the potential role of MSCs as regeneration organizers and speculate that this function emerges in CS. This concept shifts our understanding of the therapeutic mechanism underlying a widely known CS-based delivery method for regenerative medicine.
Collapse
|
13
|
Kukolj T, Trivanović D, Mojsilović S, Okić Djordjević I, Obradović H, Krstić J, Jauković A, Bugarski D. IL-33 guides osteogenesis and increases proliferation and pluripotency marker expression in dental stem cells. Cell Prolif 2018; 52:e12533. [PMID: 30430681 PMCID: PMC6430470 DOI: 10.1111/cpr.12533] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/03/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022] Open
Abstract
Objectives Soluble IL‐33 (interleukin (IL)‐1‐like cytokine) acts as endogenous alarm signal (alarmin). Since alarmins, besides activating immune system, act to restore tissue homeostasis, we investigated whether IL‐33 exerts beneficial effects on oral stem cell pull. Materials and Methods Clonogenicity, proliferation, differentiation and senescence of stem cells derived from human periodontal ligament (PDLSCs) and dental pulp (DPSCs) were determined after in vitro exposure to IL‐33. Cellular changes were detected by flow cytometry, Western blot, immunocytochemistry and semiquantitative RT‐PCR. Results IL‐33 stimulated proliferation, clonogenicity and expression of pluripotency markers, OCT‐4, SOX‐2 and NANOG, but it inhibited ALP activity and mineralization in both PDLSCs and DPSCs. Higher Ki67 expression and reduced β‐galactosidase activity in IL‐33‐treated cells were demonstrated, whereas these trends were more conspicuous in osteogenic medium. However, after 7‐day IL‐33 pretreatment, differentiation capacity of IL‐33‐pretreated cells was retained, and increased ALP activity was observed in both cell types. Results showed that IL‐33 regulates NF‐κB and β‐catenin signalling, indicating the association of these molecules with changes observed in IL‐33‐treated PDLSCs and DPSCs, particularly their proliferation, pluripotency‐associated marker expression and osteogenesis. Conclusions IL‐33 treatment impairs osteogenesis of PDLSCs and DPSCs, while increases their clonogenicity, proliferation and pluripotency marker expression. After exposure to IL‐33, osteogenic capacity of cells stayed intact. NF‐κB and β‐catenin are implicated in the effects achieved by IL‐33 in PDLSCs and DPSCs.
Collapse
Affiliation(s)
- Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Slavko Mojsilović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Ivana Okić Djordjević
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Hristina Obradović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Jelena Krstić
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
14
|
Ferracini R, Martínez Herreros I, Russo A, Casalini T, Rossi F, Perale G. Scaffolds as Structural Tools for Bone-Targeted Drug Delivery. Pharmaceutics 2018; 10:pharmaceutics10030122. [PMID: 30096765 PMCID: PMC6161191 DOI: 10.3390/pharmaceutics10030122] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
Although bone has a high potential to regenerate itself after damage and injury, the efficacious repair of large bone defects resulting from resection, trauma or non-union fractures still requires the implantation of bone grafts. Materials science, in conjunction with biotechnology, can satisfy these needs by developing artificial bones, synthetic substitutes and organ implants. In particular, recent advances in materials science have provided several innovations, underlying the increasing importance of biomaterials in this field. To address the increasing need for improved bone substitutes, tissue engineering seeks to create synthetic, three-dimensional scaffolds made from organic or inorganic materials, incorporating drugs and growth factors, to induce new bone tissue formation. This review emphasizes recent progress in materials science that allows reliable scaffolds to be synthesized for targeted drug delivery in bone regeneration, also with respect to past directions no longer considered promising. A general overview concerning modeling approaches suitable for the discussed systems is also provided.
Collapse
Affiliation(s)
- Riccardo Ferracini
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
| | - Isabel Martínez Herreros
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
| | - Antonio Russo
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
| | - Tommaso Casalini
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Vladimir-Prelog-Weg 1, 8093 Zurich, Switzerland.
- Biomaterials Laboratory, Institute for Mechanical Engineering and Materials Technology, University of Applied Sciences and Arts of Southern Switzerland, Via Cantonale 2C, Galleria, 26928 Manno, Switzerland.
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy.
| | - Giuseppe Perale
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
- Biomaterials Laboratory, Institute for Mechanical Engineering and Materials Technology, University of Applied Sciences and Arts of Southern Switzerland, Via Cantonale 2C, Galleria, 26928 Manno, Switzerland.
- Industrie Biomediche Insubri SA, Via Cantonale 67, 6805 Mezzovico-Vira, Switzerland.
| |
Collapse
|
15
|
Bunpetch V, Wu H, Zhang S, Ouyang H. From "Bench to Bedside": Current Advancement on Large-Scale Production of Mesenchymal Stem Cells. Stem Cells Dev 2018; 26:1662-1673. [PMID: 28934885 DOI: 10.1089/scd.2017.0104] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the primary cell source in cell therapy and regenerative medicine due to its extraordinary self-renewing capacity and multilineage differentiation potential. Clinical trials involving MSCs are being conducted in a range of human diseases and the number of registered cases is continuously increasing. However, a wide gap exists between the number of MSCs obtainable from the donor site and the number required for implantation to damage tissues, and also between MSC scalability and MSC phenotype stability. The clinical translation of MSCs necessitates a scalable expansion bioprocess for the biomanufacturing of therapeutically qualified cells. This review presents current achievements for expansion of MSCs. Issues involving culture condition modification, bioreactor systems, as well as microcarrier and scaffold platforms for optimal MSC systems are discussed. Most importantly, the gap between current MSC expansion and clinical application, as well as outbreak directions for the future are discussed. The present systemic review will bring new insights into future large-scale MSC expansion and clinical application.
Collapse
Affiliation(s)
- Varitsara Bunpetch
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Haoyu Wu
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Shufang Zhang
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hongwei Ouyang
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China .,4 State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou, China .,5 Department of Sports Medicine, School of Medicine, Zhejiang University , Hangzhou, China
| |
Collapse
|
16
|
Devarasetty M, Skardal A, Cowdrick K, Marini F, Soker S. Bioengineered Submucosal Organoids for In Vitro Modeling of Colorectal Cancer. Tissue Eng Part A 2018; 23:1026-1041. [PMID: 28922975 DOI: 10.1089/ten.tea.2017.0397] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The physical nature of the tumor microenvironment significantly impacts tumor growth, invasion, and response to drugs. Most in vitro tumor models are designed to study the effects of extracellular matrix (ECM) stiffness on tumor cells, while not addressing the effects of ECM's specific topography. In this study, we bioengineered submucosal organoids, using primary smooth muscle cells embedded in collagen I hydrogel, which produce aligned and parallel fiber topography similar to those found in vivo. The fiber organization in the submucosal organoids induced an epithelial phenotype in spheroids of colorectal carcinoma cells (HCT-116), which were embedded within the organoids. Conversely, unorganized fibers drove a mesenchymal phenotype in the tumor cells. HCT-116 cells in organoids with aligned fibers showed no WNT signaling activation, and conversely, WNT signaling activation was observed in organoids with disrupted fibers. Consequently, HCT-116 cells in the aligned condition exhibited decreased cellular proliferation and reduced sensitivity to 5-fluorouracil chemotherapeutic treatment compared to cells in the unorganized construct. Collectively, the results establish a unique colorectal tumor organoid model to study the effects of stromal topography on cancer cell phenotype, proliferation, and ultimately, chemotherapeutic susceptibility. In the future, such organoids can utilize patient-derived cells for precision medicine applications.
Collapse
Affiliation(s)
- Mahesh Devarasetty
- 1 Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine, Winston-Salem, North Carolina.,2 Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Aleksander Skardal
- 1 Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine, Winston-Salem, North Carolina.,2 Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine , Winston-Salem, North Carolina.,3 Comprehensive Cancer Center at Wake Forest Baptist Medical , Winston-Salem, North Carolina.,4 Department of Cancer Biology, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Kyle Cowdrick
- 1 Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Frank Marini
- 1 Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine, Winston-Salem, North Carolina.,3 Comprehensive Cancer Center at Wake Forest Baptist Medical , Winston-Salem, North Carolina.,4 Department of Cancer Biology, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Shay Soker
- 1 Wake Forest Institute for Regenerative Medicine , Wake Forest School of Medicine, Winston-Salem, North Carolina.,2 Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine , Winston-Salem, North Carolina.,3 Comprehensive Cancer Center at Wake Forest Baptist Medical , Winston-Salem, North Carolina.,4 Department of Cancer Biology, Wake Forest School of Medicine , Winston-Salem, North Carolina
| |
Collapse
|
17
|
Han S, Proctor AR, Ren J, Benoit DSW, Choe R. Temporal blood flow changes measured by diffuse correlation tomography predict murine femoral graft healing. PLoS One 2018; 13:e0197031. [PMID: 29813078 PMCID: PMC5973582 DOI: 10.1371/journal.pone.0197031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/25/2018] [Indexed: 12/18/2022] Open
Abstract
Blood flow changes during bone graft healing have the potential to provide important information about graft success, as the nutrients, oxygen, circulating cells and growth factors essential for integration are delivered by blood. However, longitudinal monitoring of blood flow changes during graft healing has been a challenge due to limitations in current techniques. To this end, non-invasive diffuse correlation tomography (DCT) was investigated to enable longitudinal monitoring of three-dimensional blood flow changes in deep tissue. Specific to this study, longitudinal blood flow changes were utilized to predict healing outcomes of common interventions for massive bone defects using a common mouse femoral defect model. Weekly blood flow changes were non-invasively measured using a diffuse correlation tomography system for 9 weeks in three types of grafts: autografts (N = 7), allografts (N = 6) and tissue-engineered allografts (N = 6). Healing outcomes were quantified using an established torsion testing method 9 weeks after transplantation. Analysis of the spatial and temporal blood flow reveals that major differences among the three groups were captured in weeks 1-5 after graft transplantation. A multivariate model to predict maximum torque by relative blood flow changes over 5 weeks after graft transplantation was built using partial least squares regression. The results reveal lower bone strength correlates with greater cumulative blood flow over an extended period of time (i.e., 1-5 weeks). The current research demonstrates that DCT-measured blood flow changes after graft transplantation can be utilized to predict long-term healing outcomes in a mouse femoral graft model.
Collapse
Affiliation(s)
- Songfeng Han
- Institute of Optics, University of Rochester, Rochester, NY, United States of America
| | - Ashley R. Proctor
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
| | - Jingxuan Ren
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Chemical Engineering, University of Rochester, Rochester, NY, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Regine Choe
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States of America
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, United States of America
- * E-mail:
| |
Collapse
|
18
|
Wang Y, Newman MR, Benoit DSW. Development of controlled drug delivery systems for bone fracture-targeted therapeutic delivery: A review. Eur J Pharm Biopharm 2018; 127:223-236. [PMID: 29471078 DOI: 10.1016/j.ejpb.2018.02.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/17/2018] [Accepted: 02/17/2018] [Indexed: 01/09/2023]
Abstract
Impaired fracture healing is a major clinical problem that can lead to patient disability, prolonged hospitalization, and significant financial burden. Although the majority of fractures heal using standard clinical practices, approximately 10% suffer from delayed unions or non-unions. A wide range of factors contribute to the risk for nonunions including internal factors, such as patient age, gender, and comorbidities, and external factors, such as the location and extent of injury. Current clinical approaches to treat nonunions include bone grafts and low-intensity pulsed ultrasound (LIPUS), which realizes clinical success only to select patients due to limitations including donor morbidities (grafts) and necessity of fracture reduction (LIPUS), respectively. To date, therapeutic approaches for bone regeneration rely heavily on protein-based growth factors such as INFUSE, an FDA-approved scaffold for delivery of bone morphogenetic protein 2 (BMP-2). Small molecule modulators and RNAi therapeutics are under development to circumvent challenges associated with traditional growth factors. While preclinical studies has shown promise, drug delivery has become a major hurdle stalling clinical translation. Therefore, this review overviews current therapies employed to stimulate fracture healing pre-clinically and clinically, including a focus on drug delivery systems for growth factors, parathyroid hormone (PTH), small molecules, and RNAi therapeutics, as well as recent advances and future promise of fracture-targeted drug delivery.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Maureen R Newman
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Chemical Engineering, 4517 Wegmans Hall, University of Rochester, Rochester, NY 14627, USA; Department of Orthopaedics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Department of Biomedical Genetics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Center for Oral Biology, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
19
|
Huang X, Zhong L, Hendriks J, Post JN, Karperien M. The Effects of the WNT-Signaling Modulators BIO and PKF118-310 on the Chondrogenic Differentiation of Human Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19020561. [PMID: 29438298 PMCID: PMC5855783 DOI: 10.3390/ijms19020561] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 01/22/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells, mainly from bone marrow, and an ideal source of cells in bone and cartilage tissue engineering. A study of the chondrogenic differentiation of MSCs is of particular interest for MSCs-based cartilage regeneration. In this study, we aimed to optimize the conditions for the chrondogenic differentiation of MSCs by regulating WNT signaling using the small molecule WNT inhibitor PKF118-310 and activator BIO. Human mesenchymal stem cells (hMSCs) were isolated from bone marrow aspirates and cultured in hMSCs proliferation medium. Pellet culture was subsequently established for three-dimensional chondrogenic differentiation of 5 weeks. WNT signaling was increased by the small molecule glycogen synthase kinase-3 inhibitor 6-bromoindirubin-3-oxim (BIO) and decreased by the WNT inhibitor PKF118-310 (PKF). The effects of BIO and PKF on the chondrogenesis of hMSCs was examined by real-time PCR, histological methods, and ELISA. We found that activation of canonical WNT-signaling by BIO significantly downregulated the expression of cartilage-specific genes SOX9, COL2A1, and ACAN, and matrix metalloproteinase genes MMP1/3/9/13, but increased ADAMTS 4/5. Inhibition of WNT signaling by PKF increased the expression of SOX9, COL2A1, ACAN, and MMP9, but decreased MMP13 and ADAMTS4/5. In addition, a high level of WNT signaling induced the expression of hypertrophic markers COL10A1, ALPL, and RUNX2, the dedifferentiation marker COL1A1, and glycolysis genes GULT1 and PGK1. Deposition of glycosaminoglycan (GAG) and collagen type II in the pellet matrix was significantly lost in the BIO-treated group and increased in the PKF-treated group. The protein level of COL10A1 was also highly induced in the BIO group. Interestingly, BIO decreased the number of apoptotic cells while PKF significantly induced apoptosis during chondrogenesis. The natural WNT antagonist DKK1 and the protein level of MMP1 in the pellet culture medium were decreased after PKF treatment. All of these chondrogenic effects appeared to be mediated through the canonical WNT signaling pathway, since the target gene Axin2 and other WNT members, such as TCF4 and β-catenin, were upregulated by BIO and downregulated by PKF, respectively, and BIO induced nuclear translocation of β-catenin while PKF inhibited β-catenin translocation into the nucleus. We concluded that addition of BIO to a chondrogenic medium of hMSCs resulted in a loss of cartilage formation, while PKF induced chondrogenic differentiation and cartilage matrix deposition and inhibited hypertrophic differentiation. However, BIO promoted cell survival by inhibiting apoptosis while PKF induced cell apoptosis. This result indicates that either an overexpression or overinhibition of WNT signaling to some extent causes harmful effects on chondrogenic differentiation. Cartilage tissue engineering could benefit from the adjustment of the critical level of WNT signaling during chondrogenesis of hMSC.
Collapse
Affiliation(s)
- Xiaobin Huang
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Leilei Zhong
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Jan Hendriks
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Janine N Post
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Marcel Karperien
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| |
Collapse
|
20
|
Bunpetch V, Zhang ZY, Zhang X, Han S, Zongyou P, Wu H, Hong-Wei O. Strategies for MSC expansion and MSC-based microtissue for bone regeneration. Biomaterials 2017; 196:67-79. [PMID: 29602560 DOI: 10.1016/j.biomaterials.2017.11.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/31/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) have gained increasing attention as a potential approach for the treatment of bone injuries due to their multi-lineage differentiation potential and also their ability to recognize and home to damaged tissue sites, secreting bioactive factors that can modulate the immune system and enhance tissue repair. However, a wide gap between the number of MSCs obtainable from the donor site and the number required for implantation, as well as the lack of understanding of MSC functions under different in vitro and in vivo microenvironment, hinders the progression of MSCs toward clinical settings. The clinical translation of MSCs pre-requisites a scalable expansion process for the biomanufacturing of therapeutically qualified cells. This review briefly introduces the features of implanted MSCs to determine the best strategies to optimize their regenerative capacity, as well as the current MSC implantation for bone diseases. Current achievements for expansion of MSCs using various culturing methods, bioreactor technologies, biomaterial platforms, as well as microtissue-based expansion strategies are also discussed, providing new insights into future large-scale MSC expansion and clinical applications.
Collapse
Affiliation(s)
- Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, China; Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing Technologies of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, 510150, China.
| | - Xiaoan Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, China; Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shan Han
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, China; Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pan Zongyou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, China; Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haoyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, China; Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ouyang Hong-Wei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, China; Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China; Department of Sports Medicine, School of Medicine, Zhejiang University, China; Translational Research Centre of Regenerative Medicine and 3D Printing Technologies of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, No.63 Duobao Road, Liwan District, Guangzhou City, Guangdong Province, 510150, China.
| |
Collapse
|
21
|
Wang Y, Newman MR, Ackun-Farmmer M, Baranello MP, Sheu TJ, Puzas JE, Benoit DSW. Fracture-Targeted Delivery of β-Catenin Agonists via Peptide-Functionalized Nanoparticles Augments Fracture Healing. ACS NANO 2017; 11:9445-9458. [PMID: 28881139 PMCID: PMC5736386 DOI: 10.1021/acsnano.7b05103] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Despite several decades of progress, bone-specific drug delivery is still a major challenge. Current bone-acting drugs require high-dose systemic administration which decreases therapeutic efficacy and increases off-target tissue effects. Here, a bone-targeted nanoparticle (NP) delivery system for a β-catenin agonist, 3-amino-6-(4-((4-methylpiperazin-1-yl)sulfonyl)phenyl)-N-(pyridin-3-yl)pyrazine-2-carboxamide, a glycogen synthase kinase 3 beta (GSK-3β) inhibitor, was developed to enhance fracture healing. The GSK-3β inhibitor loading capacity was found to be 15 wt % within highly stable poly(styrene-alt-maleic anhydride)-b-poly(styrene) NPs, resulting in ∼50 nm particles with ∼ -30 mV surface charge. A peptide with high affinity for tartrate-resistant acid phosphatase (TRAP), a protein deposited by osteoclasts on bone resorptive surfaces, was introduced to the NP corona to achieve preferential delivery to fractured bone. Targeted NPs showed improved pharmacokinetic profiles with greater accumulation at fractured bone, accompanied by significant uptake in regenerative cell types (mesenchymal stem cells (MSCs) and osteoblasts). MSCs treated with drug-loaded NPs in vitro exhibited 2-fold greater β-catenin signaling than free drug that was sustained for 5 days. To verify similar activity in vivo, TOPGAL reporter mice bearing fractures were treated with targeted GSK-3β inhibitor-loaded NPs. Robust β-galactosidase activity was observed in fracture callus and periosteum treated with targeted carriers versus controls, indicating potent β-catenin activation during the healing process. Enhanced bone formation and microarchitecture were observed in mice treated with GSK-3β inhibitor delivered via TRAP-binding peptide-targeted NPs. Specifically, increased bone bridging, ∼4-fold greater torsional rigidity, and greater volumes of newly deposited bone were observed 28 days after treatment, indicating expedited fracture healing.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen R. Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Marian Ackun-Farmmer
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Michael P. Baranello
- Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States
| | - Tzong-Jen Sheu
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - J. Edward Puzas
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States
- Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
22
|
Mills KM, Szczerkowski JLA, Habib SJ. Wnt ligand presentation and reception: from the stem cell niche to tissue engineering. Open Biol 2017; 7:rsob.170140. [PMID: 28814649 PMCID: PMC5577451 DOI: 10.1098/rsob.170140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cells reside in niches where spatially restricted signals maintain a delicate balance between stem cell self-renewal and differentiation. Wnt family proteins are particularly suited for this role as they are modified by lipids, which constrain and spatially regulate their signalling range. In recent years, Wnt/β-catenin signalling has been shown to be essential for the self-renewal of a variety of mammalian stem cells. In this review, we discuss Wnt-responsive stem cells in their niche, and mechanisms by which Wnt ligands are presented to responsive cells. We also highlight recent progress in molecular visualization that has allowed for the monitoring of Wnt signalling within the stem cell compartment and new approaches to recapitulate this niche signalling in vitro Indeed, new technologies that present Wnt in a localized manner and mimic the three-dimensional microenvironment of stem cells will advance our understanding of Wnt signalling in the stem cell niche. These advances will expand current horizons to exploit Wnt ligands in the rapidly evolving fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Kate M Mills
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - James L A Szczerkowski
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| |
Collapse
|
23
|
Lowndes M, Junyent S, Habib SJ. Constructing cellular niche properties by localized presentation of Wnt proteins on synthetic surfaces. Nat Protoc 2017; 12:1498-1512. [PMID: 28686585 DOI: 10.1038/nprot.2017.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Wnt signaling is crucial during embryonic development and for the maintenance of adult tissues. Depending on the tissue type, the Wnt pathway can promote stem cell self-renewal and/or direct lineage commitment. Wnt proteins are subject to lipid modification, often restricting them to act in a localized manner on responsive cells. Most methods for inducing Wnt signaling in stem cell cultures do not control the spatial presentation of the protein. To recreate the local presentation of Wnt proteins often seen in vivo, we previously developed a method to immobilize the protein onto synthetic surfaces. Here we describe a detailed protocol based on covalent binding of nucleophilic groups on Wnt proteins to activated carboxylic acid (COOH) or glutaraldehyde (COH) groups functionalized on synthetic surfaces. As an example, we describe how this method can be used to covalently immobilize Wnt3a proteins on microbeads or a glass surface. This procedure requires ∼3 h and allows for the hydrophobic protein to be stored in the absence of detergent. The immobilization efficiency of active Wnt proteins can be assessed using different T-cell factor (TCF) reporter assays as a readout for Wnt/β-catenin-dependent transcription. Immobilization efficiency can be measured 12-18 h after seeding the cells and takes 2-4 h. The covalent immobilization of Wnt proteins can also be used for single-cell analysis using Wnt-coated microbeads (12-18 h of live imaging) and to create a Wnt platform on a glass surface for stem cell maintenance and cell population analysis (3 d). The simple chemistry used for Wnt immobilization allows for adaptation to new materials and other developmental signals. Therefore, this method can also be incorporated into tissue engineering platforms in which depletion of the stem cell pool restricts the complexity and maturity of the tissue developed.
Collapse
Affiliation(s)
- Molly Lowndes
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Sergi Junyent
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| |
Collapse
|
24
|
Sato S, Tang YJ, Wei Q, Hirata M, Weng A, Han I, Okawa A, Takeda S, Whetstone H, Nadesan P, Kirsch DG, Wunder JS, Alman BA. Mesenchymal Tumors Can Derive from Ng2/Cspg4-Expressing Pericytes with β-Catenin Modulating the Neoplastic Phenotype. Cell Rep 2016; 16:917-927. [PMID: 27425618 PMCID: PMC4963269 DOI: 10.1016/j.celrep.2016.06.058] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 05/02/2016] [Accepted: 06/11/2016] [Indexed: 02/07/2023] Open
Abstract
The cell of origin for most mesenchymal tumors is unclear. One cell type that contributes to this lineages is the pericyte, a cell expressing Ng2/Cspg4. Using lineage tracing, we demonstrated that bone and soft tissue sarcomas driven by the deletion of the Trp53 tumor suppressor, or desmoid tumors driven by a mutation in Apc, can derive from cells expressing Ng2/Cspg4. Deletion of the Trp53 tumor suppressor gene in these cells resulted in the bone and soft tissue sarcomas that closely resemble human sarcomas, while stabilizing β-catenin in this same cell type caused desmoid tumors. Comparing expression between Ng2/Cspg4-expressing pericytes lacking Trp53 and sarcomas that arose from deletion of Trp53 showed inhibition of β-catenin signaling in the sarcomas. Activation of β-catenin inhibited the formation and growth of sarcomas. Thus, pericytes can be a cell of origin for mesenchymal tumors, and β-catenin dysregulation plays an important role in the neoplastic phenotype.
Collapse
Affiliation(s)
- Shingo Sato
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada; Department of Orthopaedic Surgery, Tokyo Medical and Dental, University Graduate School and Faculty of Medicine, Tokyo 113-8510, Japan; Department of Physiology and Cell Biology, Graduate School and Faculty of Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yuning J Tang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA
| | - Qingxia Wei
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| | - Makoto Hirata
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| | - Angela Weng
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| | - Ilkyu Han
- Department of Orthopaedic Surgery, Seoul National University Hospital, Seoul 151-742, Republic of Korea
| | - Atsushi Okawa
- Department of Orthopaedic Surgery, Tokyo Medical and Dental, University Graduate School and Faculty of Medicine, Tokyo 113-8510, Japan
| | - Shu Takeda
- Department of Physiology and Cell Biology, Graduate School and Faculty of Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Heather Whetstone
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| | - Puvindran Nadesan
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Jay S Wunder
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Benjamin A Alman
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
25
|
Langhans MT, Yu S, Tuan RS. Stem Cells in Skeletal Tissue Engineering: Technologies and Models. Curr Stem Cell Res Ther 2016; 11:453-474. [PMID: 26423296 DOI: 10.2174/1574888x10666151001115248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
This review surveys the use of pluripotent and multipotent stem cells in skeletal tissue engineering. Specific emphasis is focused on evaluating the function and activities of these cells in the context of development in vivo, and how technologies and methods of stem cell-based tissue engineering for stem cells must draw inspiration from developmental biology. Information on the embryonic origin and in vivo differentiation of skeletal tissues is first reviewed, to shed light on the persistence and activities of adult stem cells that remain in skeletal tissues after embryogenesis. Next, the development and differentiation of pluripotent stem cells is discussed, and some of their advantages and disadvantages in the context of tissue engineering are presented. The final section highlights current use of multipotent adult mesenchymal stem cells, reviewing their origin, differentiation capacity, and potential applications to tissue engineering.
Collapse
Affiliation(s)
| | | | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219, USA.
| |
Collapse
|
26
|
Newman MR, Benoit DS. Local and targeted drug delivery for bone regeneration. Curr Opin Biotechnol 2016; 40:125-132. [PMID: 27064433 DOI: 10.1016/j.copbio.2016.02.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 01/08/2023]
Abstract
While experimental bone regeneration approaches commonly employ cells, technological hurdles prevent translation of these therapies. Alternatively, emulating the spatiotemporal cascade of endogenous factors through controlled drug delivery may provide superior bone regenerative approaches. Surgically placed drug depots have clinical indications. Additionally, noninvasive systemic delivery can be used as needed for poorly healing bone injuries. However, a major hurdle for systemic delivery is poor bone biodistribution of drugs. Thus, peptides, aptamers, and phosphate-rich compounds with specificity toward proteins, cells, and molecules within the regenerative bone microenvironment may enable the design of targeted carriers with bone biodistribution greater than that achieved by drug alone. These carriers, combined with osteoregenerative drugs and/or stimuli-sensitive linkers, may enhance bone regeneration while minimizing off-target tissue effects.
Collapse
Affiliation(s)
- Maureen R Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Danielle Sw Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA; Department of Chemical Engineering, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
27
|
Gao B, Huang Q, Jie Q, Lu WG, Wang L, Li XJ, Sun Z, Hu YQ, Chen L, Liu BH, Liu J, Yang L, Luo ZJ. GPR120: A bi-potential mediator to modulate the osteogenic and adipogenic differentiation of BMMSCs. Sci Rep 2015; 5:14080. [PMID: 26365922 PMCID: PMC4568495 DOI: 10.1038/srep14080] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/18/2015] [Indexed: 12/14/2022] Open
Abstract
Free fatty acids display diverse effects as signalling molecules through GPCRs in addition to their involvement in cellular metabolism. GPR120, a G protein-coupled receptor for long-chain unsaturated fatty acids, has been reported to mediate adipogenesis in lipid metabolism. However, whether GPR120 also mediates osteogenesis and regulates BMMSCs remain unclear. In this study, we showed that GPR120 targeted the bi-potential differentiation of BMMSCs in a ligand dose-dependent manner. High concentrations of TUG-891 (a highly selective agonist of GPR120) promoted osteogenesis via the Ras-ERK1/2 cascade, while low concentrations elevated P38 and increased adipogenesis. The fine molecular regulation of GPR120 was implemented by up-regulating different integrin subunits (α1, α2 and β1; α5 and β3). The administration of high doses of TUG-891 rescued oestrogen-deficient bone loss in vivo, further supporting an essential role of GPR120 in bone metabolism. Our findings, for the first time, showed that GPR120-mediated cellular signalling determines the bi-potential differentiation of BMMSCs in a dose-dependent manner. Additionally, the induction of different integrin subunits was involved in the cytoplasmic regulation of a seesaw-like balance between ERK and p38 phosphorylation. These findings provide new hope for developing novel remedies to treat osteoporosis by adjusting the GPR120-mediated differentiation balance of BMMSCs.
Collapse
Affiliation(s)
- Bo Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Qiang Huang
- Lanzhou General Hospital of Lanzhou Military Command, Lanzhou Gansu, 730050, People's Republic of China
| | - Qiang Jie
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Wei-Guang Lu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Long Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xiao-Jie Li
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Zhen Sun
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Ya-Qian Hu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Li Chen
- KMEB, Molecular Endocrinology, Campusvej 55, DK-5230 Odense M, Denmark
| | - Bao-Hua Liu
- Health Science Center, Shenzhen University, 3688 Nanhai Ave, Shenzhen 518060, People's Republic of China
| | - Jian Liu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Zhuo-Jing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
28
|
Emulating native periosteum cell population and subsequent paracrine factor production to promote tissue engineered periosteum-mediated allograft healing. Biomaterials 2015; 52:426-40. [PMID: 25818449 DOI: 10.1016/j.biomaterials.2015.02.064] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/10/2015] [Accepted: 02/13/2015] [Indexed: 01/01/2023]
Abstract
Emulating autograft healing within the context of decellularized bone allografts has immediate clinical applications in the treatment of critical-sized bone defects. The periosteum, a thin, osteogenic tissue that surrounds bone, houses a heterogenous population of stem cells and osteoprogenitors. There is evidence that periosteum-cell derived paracrine factors, specifically vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP2), orchestrate autograft healing through host cell recruitment and subsequent tissue elaboration. In previous work, we demonstrated that the use of poly(ethylene glycol) (PEG) hydrogels as a tissue engineered (T.E.) periosteum to localize mesenchymal stem cells (MSCs) to the surface of decellularized bone enhances allograft healing and integration. Herein, we utilize a mixed population of 50:50 MSCs and osteoprogenitor cells to better mimic native periosteum cell population and paracrine factor production to further promote allograft healing. This mixed cell population was localized to the surface of decellularized allografts within degradable hydrogels and shown to expedite allograft healing. Specifically, bone callus formation and biomechanical graft-host integration are increased as compared to unmodified allografts. These results demonstrate the dual importance of periosteum-mediated paracrine factors orchestrating host cell recruitment as well as new bone formation while developing clinically translatable strategies for allograft healing and integration.
Collapse
|
29
|
Chen AX, Hoffman MD, Chen CS, Shubin AD, Reynolds DS, Benoit DSW. Disruption of cell-cell contact-mediated notch signaling via hydrogel encapsulation reduces mesenchymal stem cell chondrogenic potential: winner of the Society for Biomaterials Student Award in the Undergraduate Category, Charlotte, NC, April 15 to 18, 2015. J Biomed Mater Res A 2014; 103:1291-302. [PMID: 25504509 DOI: 10.1002/jbm.a.35383] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/17/2014] [Accepted: 12/04/2014] [Indexed: 12/20/2022]
Abstract
Cell-cell contact-mediated Notch signaling is essential for mesenchymal stem cell (MSC) chondrogenesis during development. However, subsequent deactivation of Notch signaling is also required to allow for stem cell chondrogenic progression. Recent literature has shown that Notch signaling can also influence Wnt/β-catenin signaling, critical for MSC differentiation, through perturbations in cell-cell contacts. Traditionally, abundant cell-cell contacts, consistent with development, are emulated in vitro using pellet cultures for chondrogenesis. However, cells are often encapsulated within biomaterials-based scaffolds, such as hydrogels, to improve therapeutic cell localization in vivo. To explore the role of Notch and Wnt/β-catenin signaling in the context of hydrogel-encapsulated MSC chondrogenesis, we compared signaling and differentiation capacity of MSCs in both hydrogels and traditional pellet cultures. We demonstrate that encapsulation within poly(ethylene glycol) hydrogels reduces cell-cell contacts, and both Notch (7.5-fold) and Wnt/β-catenin (84.7-fold) pathway activation. Finally, we demonstrate that following establishment of cell-cell contacts and transient Notch signaling in pellet cultures, followed by Notch signaling deactivation, resulted in a 1.5-fold increase in MSC chondrogenesis. Taken together, these findings support that cellular condensation, and establishment of initial cell-cell contacts is critical for MSC chondrogenesis, and this process is inhibited by hydrogel encapsulation.
Collapse
Affiliation(s)
- Amanda X Chen
- Department of Biomedical Engineering, University of Rochester, 207 Robert B. Goergen Hall, Box 270168, Rochester, New York, 14627-0168
| | | | | | | | | | | |
Collapse
|
30
|
Dong Y, Long T, Wang C, Mirando AJ, Chen J, O'Keefe RJ, Hilton MJ. NOTCH-Mediated Maintenance and Expansion of Human Bone Marrow Stromal/Stem Cells: A Technology Designed for Orthopedic Regenerative Medicine. Stem Cells Transl Med 2014; 3:1456-66. [PMID: 25368376 DOI: 10.5966/sctm.2014-0034] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human bone marrow-derived stromal/stem cells (BMSCs) have great therapeutic potential for treating skeletal disease and facilitating skeletal repair, although maintaining their multipotency and expanding these cells ex vivo have proven difficult. Because most stem cell-based applications to skeletal regeneration and repair in the clinic would require large numbers of functional BMSCs, recent research has focused on methods for the appropriate selection, expansion, and maintenance of BMSC populations during long-term culture. We describe here a novel biological method that entails selection of human BMSCs based on NOTCH2 expression and activation of the NOTCH signaling pathway in cultured BMSCs via a tissue culture plate coated with recombinant human JAGGED1 (JAG1) ligand. We demonstrate that transient JAG1-mediated NOTCH signaling promotes human BMSC maintenance and expansion while increasing their skeletogenic differentiation capacity, both ex vivo and in vivo. This study is the first of its kind to describe a NOTCH-mediated methodology for the maintenance and expansion of human BMSCs and will serve as a platform for future clinical or translational studies aimed at skeletal regeneration and repair.
Collapse
Affiliation(s)
- Yufeng Dong
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| | - Teng Long
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| | - Cuicui Wang
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| | - Anthony J Mirando
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jianquan Chen
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| | - Regis J O'Keefe
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthew J Hilton
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, and Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA; Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA; Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
31
|
Degradable hydrogels for spatiotemporal control of mesenchymal stem cells localized at decellularized bone allografts. Acta Biomater 2014; 10:3431-41. [PMID: 24751534 DOI: 10.1016/j.actbio.2014.04.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/04/2014] [Accepted: 04/11/2014] [Indexed: 12/18/2022]
Abstract
The transplantation of cells, such as mesenchymal stem cells (MSCs), has numerous applications in the field of regenerative medicine. For cell transplantation strategies to be successful therapeutically, cellular localization and persistence must be controlled to maximize cell-mediated contributions to healing. Herein, we demonstrate that hydrolytic degradation of poly(ethylene glycol) (PEG) hydrogels can be used to spatiotemporally control encapsulated MSC localization to decellularized bone allografts, both in vitro and in vivo. By altering the number of hydrolytically degradable lactide repeat units within PEG-d,l-lactide-methacrylate macromers, a series of hydrogels was synthesized that degraded over ∼1, 2 and 3weeks. MSCs were encapsulated within these hydrogels formed around decellularized bone allografts, and non-invasive, longitudinal fluorescence imaging was used to track cell persistence both in vitro and in vivo. Spatiotemporal localization of MSCs to the exterior of bone allograft surfaces was similar to in vitro hydrogel degradation kinetics despite hydrogel mesh sizes being ∼2-3 orders of magnitude smaller than MSC size throughout the degradation process. Thus, localized, cell-mediated degradation and MSC migration from the hydrogels are suspected, particularly as ∼10% of the total transplanted MSC population was shown to persist in close proximity (within ∼650μm) to grafts 7weeks after complete hydrogel degradation. This work demonstrates the therapeutic utility of PEG-based hydrogels for controlling spatiotemporal cell transplantation for a myriad of regenerative medicine strategies.
Collapse
|
32
|
Pavlaki K, Pontikoglou CG, Demetriadou A, Batsali AK, Damianaki A, Simantirakis E, Kontakis M, Galanopoulos A, Kotsianidis I, Kastrinaki MC, Papadaki HA. Impaired proliferative potential of bone marrow mesenchymal stromal cells in patients with myelodysplastic syndromes is associated with abnormal WNT signaling pathway. Stem Cells Dev 2014; 23:1568-81. [PMID: 24617415 DOI: 10.1089/scd.2013.0283] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It has been shown that bone marrow mesenchymal stromal cells (MSCs) from patients with myelodysplastic syndromes (MDSs) display defective proliferative potential. We have probed the impaired replicative capacity of culture-expanded MSCs in MDS patients (n=30) compared with healthy subjects (n=32) by studying senescence characteristics and gene expression associated with WNT/transforming growth factor-β1 (TGFB1) signaling pathways. We have also explored the consequences of the impaired patient MSC proliferative potential by investigating their differentiation potential and the capacity to support normal CD34(+) cell growth under coculture conditions. Patient MSCs displayed decreased gene expression of the senescence-associated cyclin-dependent kinase inhibitors CDKN1A, CDKN2A, and CDKN2B, along with PARG1, whereas the mean telomere length was upregulated in patient MSCs. MDS-derived MSCs exhibited impaired capacity to support normal CD34(+) myeloid and erythroid colony formation. No significant changes were observed between patients and controls in gene expression related to TGFB1 pathway. Patient MSCs displayed upregulated non-canonical WNT expression, combined with downregulated canonical WNT expression and upregulated canonical WNT inhibitors. MDS-derived MSCs displayed defective osteogenic and adipogenic lineage priming under non-differentiating culture conditions. Pharmacological activation of canonical WNT signaling in patient MDSs led to an increase in cell proliferation and upregulation in the expression of early osteogenesis-related genes. This study indicates abnormal WNT signaling in MSCs of MDS patients and supports the concept of a primary MSC defect that might have a contributory effect in MDS natural history.
Collapse
Affiliation(s)
- Konstantia Pavlaki
- 1 Department of Haematology, University of Crete School of Medicine , Heraklion, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cai SX, Liu AR, He HL, Chen QH, Yang Y, Guo FM, Huang YZ, Liu L, Qiu HB. Stable Genetic Alterations of β-Catenin and ROR2 Regulate the Wnt Pathway, Affect the Fate of MSCs. J Cell Physiol 2014; 229:791-800. [PMID: 24590964 DOI: 10.1002/jcp.24500] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/17/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Shi-Xia Cai
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Ai-Ran Liu
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Hong-Li He
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Qi-Hong Chen
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Yi Yang
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Feng-Mei Guo
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Ying-Zi Huang
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Ling Liu
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| | - Hai-Bo Qiu
- Department of Critical Care Medicine; Nanjing Zhong-da Hospital, School of Medicine; Southeast University; Nanjing P.R. China
| |
Collapse
|
34
|
Van Hove AH, Wilson BD, Benoit DSW. Microwave-assisted functionalization of poly(ethylene glycol) and on-resin peptides for use in chain polymerizations and hydrogel formation. J Vis Exp 2013:e50890. [PMID: 24193366 PMCID: PMC3968890 DOI: 10.3791/50890] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
One of the main benefits to using poly(ethylene glycol) (PEG) macromers in hydrogel formation is synthetic versatility. The ability to draw from a large variety of PEG molecular weights and configurations (arm number, arm length, and branching pattern) affords researchers tight control over resulting hydrogel structures and properties, including Young’s modulus and mesh size. This video will illustrate a rapid, efficient, solvent-free, microwave-assisted method to methacrylate PEG precursors into poly(ethylene glycol) dimethacrylate (PEGDM). This synthetic method provides much-needed starting materials for applications in drug delivery and regenerative medicine. The demonstrated method is superior to traditional methacrylation methods as it is significantly faster and simpler, as well as more economical and environmentally friendly, using smaller amounts of reagents and solvents. We will also demonstrate an adaptation of this technique for on-resin methacrylamide functionalization of peptides. This on-resin method allows the N-terminus of peptides to be functionalized with methacrylamide groups prior to deprotection and cleavage from resin. This allows for selective addition of methacrylamide groups to the N-termini of the peptides while amino acids with reactive side groups (e.g. primary amine of lysine, primary alcohol of serine, secondary alcohols of threonine, and phenol of tyrosine) remain protected, preventing functionalization at multiple sites. This article will detail common analytical methods (proton Nuclear Magnetic Resonance spectroscopy (;H-NMR) and Matrix Assisted Laser Desorption Ionization Time of Flight mass spectrometry (MALDI-ToF)) to assess the efficiency of the functionalizations. Common pitfalls and suggested troubleshooting methods will be addressed, as will modifications of the technique which can be used to further tune macromer functionality and resulting hydrogel physical and chemical properties. Use of synthesized products for the formation of hydrogels for drug delivery and cell-material interaction studies will be demonstrated, with particular attention paid to modifying hydrogel composition to affect mesh size, controlling hydrogel stiffness and drug release.
Collapse
Affiliation(s)
- Amy H Van Hove
- Department of Biomedical Engineering, University of Rochester
| | | | | |
Collapse
|
35
|
The effect of mesenchymal stem cells delivered via hydrogel-based tissue engineered periosteum on bone allograft healing. Biomaterials 2013; 34:8887-98. [PMID: 23958029 DOI: 10.1016/j.biomaterials.2013.08.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/01/2013] [Indexed: 12/18/2022]
Abstract
Allografts remain the clinical "gold standard" for treatment of critical sized bone defects despite minimal engraftment and ∼60% long-term failure rates. Therefore, the development of strategies to improve allograft healing and integration are necessary. The periosteum and its associated stem cell population, which are lacking in allografts, coordinate autograft healing. Herein we utilized hydrolytically degradable hydrogels to transplant and localize mesenchymal stem cells (MSCs) to allograft surfaces, creating a periosteum mimetic, termed a 'tissue engineered periosteum'. Our results demonstrated that this tissue engineering approach resulted in increased graft vascularization (∼2.4-fold), endochondral bone formation (∼2.8-fold), and biomechanical strength (1.8-fold), as compared to untreated allografts, over 16 weeks of healing. Despite this enhancement in healing, the process of endochondral ossification was delayed compared to autografts, requiring further modifications for this approach to be clinically acceptable. However, this bottom-up biomaterials approach, the engineered periosteum, can be augmented with alternative cell types, matrix cues, growth factors, and/or other small molecule drugs to expedite the process of ossification.
Collapse
|