1
|
Islam F, Javdan SB, Lewis MR, Craig JD, Wu H, Deans TL. Programming megakaryocytes to produce engineered platelets for delivering non-native proteins. Commun Biol 2025; 8:638. [PMID: 40253534 PMCID: PMC12009418 DOI: 10.1038/s42003-025-08017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/28/2025] [Indexed: 04/21/2025] Open
Abstract
Platelets are anucleate cells naturally filled with secretory granules that store large amounts of protein to be released in response to certain physiological conditions. Cell engineering can endow platelets with the ability to deliver non-native proteins by modifying them as they develop during the cell fate process. This study presents a strategy to efficiently generate mouse platelets from pluripotent stem cells and demonstrates their potential as bioengineered protein delivery platforms. By modifying megakaryocytes, the progenitor cells of platelets, we successfully engineered platelets capable of packaging and delivering non-native proteins. These engineered platelets can offer flexible delivery platforms to release non-native proteins in a controlled manner upon activation when packaged into α-granules or deliver active enzymes to genetically alter recipient cells. Our findings highlight platelets as a promising tool for protein delivery in cell therapy applications.
Collapse
Affiliation(s)
- Farhana Islam
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Shwan B Javdan
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Mitchell R Lewis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - James D Craig
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Han Wu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Tara L Deans
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Cimino A, Pat F, Oyebamiji O, Pham CTN, Herzog ED, Guilak F. Dual-responsive synthetic gene circuit for dynamic biologic drug delivery via inflammatory and circadian signaling pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644403. [PMID: 40166199 PMCID: PMC11957123 DOI: 10.1101/2025.03.20.644403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Engineered cells provide versatile tools for precise, tunable drug delivery, especially when synthetic stimulus-responsive gene circuits are incorporated. In many complex disease conditions, endogenous pathologic signals such as inflammation can vary dynamically over different time scales. For example, in autoimmune conditions such as rheumatoid arthritis or juvenile idiopathic arthritis, local (joint) and systemic inflammatory signals fluctuate daily, peaking in the early morning, but can also persist over long periods of time, triggering flare-ups that can last weeks to months. However, treatment with disease-modifying anti-rheumatic drugs is typically provided at continuous high doses, regardless of disease activity and without consideration for levels of inflammatory signals. In previous studies, we have developed cell-based drug delivery systems that can automatically address the different scales of flares using either chronogenetic circuits (i.e., clock gene-responsive elements) that can be tuned for optimal drug delivery to dampen circadian variations in inflammatory levels or inflammation-responsive circuits (i.e., NF-κB-sensitive elements) that can respond to sustained arthritis flares on demand with proportional synthesis of drug. The goal of this study was to develop a novel dual-responsive synthetic gene circuit that responds to both circadian and inflammatory inputs using OR-gate logic for both daily timed therapeutic output and enhanced therapeutic output during chronic inflammatory conditions. Results We developed a synthetic gene circuit driven by tandem inflammatory NF-κB and circadian E'-box response elements. When engineered into induced pluripotent stem cells that were chondrogenically differentiated, the gene circuit demonstrated basal-level circadian output with enhanced stimulus-responsive output during an inflammatory challenge shown by bioluminescence monitoring. Similarly, this system exhibited enhanced therapeutic levels of biologic drug interleukin-1 receptor antagonist (IL-1Ra) during an inflammatory challenge in differentiated cartilage pellets. This dual-responsive therapeutic gene circuit mitigated both the inflammatory response as measured by bioluminescence reporter output and tissue-level degradation during conditions mimicking an arthritic flare. Conclusions The dual-responsive synthetic gene circuit developed herein responds to input cues from two key homeostatic transcriptional networks, enabling dynamic and tunable output. This proof-of-concept approach has the potential to match drug delivery to disease activity for optimal outcomes that addresses the complex environment of inflammatory arthritis.
Collapse
|
3
|
Cimino A, Pat F, Oyebamiji O, Pferdehirt L, Pham CTN, Herzog ED, Guilak F. Programmable chronogenetic gene circuits for self-regulated circadian delivery of biologic drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643274. [PMID: 40161636 PMCID: PMC11952517 DOI: 10.1101/2025.03.14.643274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cells of the body rely on the circadian clock to orchestrate daily changes in physiology that impact both homeostatic and pathological conditions, such as the inflammatory autoimmune disease rheumatoid arthritis (RA). In RA, high levels of proinflammatory cytokines peak early in the morning hours, reflected by daily changes in joint stiffness. Chronotherapy (or circadian medicine) seeks to delivery drugs at optimal times to maximize their efficacy. However, chronotherapy remains a largely unexplored approach for disease modifying, antirheumatic treatment, particularly for cell-based therapies. In this study, we developed autonomous chronogenetic gene circuits that produce the biologic drug interleukin-1 receptor antagonist (IL-1Ra) with desired phase and amplitude. We compared expression of IL-1Ra from circuits that contained different circadian promoter elements (E'-boxes, D-boxes, or RREs) and their ability to respond to inflammatory challenges in murine pre-differentiated induced pluripotent stem cells (PDiPSC) or engineered cartilage pellets. We confirmed that each circuit reliably peaked at a distinct circadian time over multiple days. Engineered cells generated significant amounts of IL-1Ra on a circadian basis, which protected them from circadian dysregulation and inflammatory damage. These programmable chronogenetic circuits have the potential to align with an individual's circadian rhythm for optimized, self-regulated daily drug delivery.
Collapse
|
4
|
Pferdehirt L, Damato AR, Lenz KL, Gonzalez-Aponte MF, Palmer D, Meng QJ, Herzog ED, Guilak F. A synthetic chronogenetic gene circuit for programmed circadian drug delivery. Nat Commun 2025; 16:1457. [PMID: 39920119 PMCID: PMC11806060 DOI: 10.1038/s41467-025-56584-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
Circadian medicine, the delivery of therapeutic interventions based on an individual's daily rhythms, has shown improved efficacy and reduced side-effects for various treatments. Rheumatoid arthritis and other inflammatory diseases are characterized by diurnal changes in cytokines, leading to inflammatory flares, with peak disease activity in the early morning. Using a combination of synthetic biology and tissue engineering, we developed circadian-based gene circuits, termed "chronogenetics", that express a prescribed transgene downstream of the core clock gene promoter, Period2 (Per2). Gene circuits were transduced into induced pluripotent stem cells that were tissue-engineered into cartilage constructs. Our anti-inflammatory chronogenetic constructs produced therapeutic concentrations of interleukin-1 receptor antagonist in vitro. Once implanted in vivo, the constructs expressed circadian rhythms and entrained to daily light cycles, producing daily increases in biologic drug at the peak of Per2 expression. This approach represents the development of a cell-based chronogenetic therapy for various applications in circadian medicine.
Collapse
Affiliation(s)
- Lara Pferdehirt
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children - Saint Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Anna R Damato
- Department of Biology, Washington University, St. Louis, MO, USA
| | - Kristin L Lenz
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children - Saint Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Daniel Palmer
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children - Saint Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Qing-Jun Meng
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Erik D Herzog
- Department of Biology, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Shriners Hospitals for Children - Saint Louis, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
| |
Collapse
|
5
|
Klimak M, Cimino A, Lenz KL, Springer LE, Collins KH, Harasymowicz NS, Xu N, Pham CTN, Guilak F. Engineered self-regulating macrophages for targeted anti-inflammatory drug delivery. Arthritis Res Ther 2024; 26:190. [PMID: 39501398 PMCID: PMC11539832 DOI: 10.1186/s13075-024-03425-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by increased levels of inflammation that primarily manifests in the joints. Macrophages act as key drivers for the progression of RA, contributing to the perpetuation of chronic inflammation and dysregulation of pro-inflammatory cytokines such as interleukin 1 (IL-1). The goal of this study was to develop a macrophage-based cell therapy for biologic drug delivery in an autoregulated manner. METHODS For proof-of-concept, we developed "smart" macrophages to mitigate the effects of IL-1 by delivering its inhibitor, IL-1 receptor antagonist (IL-1Ra). Bone marrow-derived macrophages were lentivirally transduced with a synthetic gene circuit that uses an NF-κB inducible promoter upstream of either the Il1rn or firefly luciferase transgenes. Two types of joint like cells were utilized to examine therapeutic protection in vitro, miPSCs derived cartilage and isolated primary mouse synovial fibroblasts while the K/BxN mouse model of RA was utilized to examine in vivo therapeutic protection. RESULTS These engineered macrophages were able to repeatably produce therapeutic levels of IL-1Ra that could successfully mitigate inflammatory activation in co-culture with both tissue-engineered cartilage constructs and synovial fibroblasts. Following injection in vivo, macrophages homed to sites of inflammation and mitigated disease severity in the K/BxN mouse model of RA. CONCLUSION These findings demonstrate the successful development of engineered macrophages that possess the ability for controlled, autoregulated production of IL-1 based on inflammatory signaling such as via the NF-κB pathway to mitigate the effects of this cytokine for applications in RA or other inflammatory diseases. This system provides proof of concept for applications in other immune cell types as self-regulating delivery systems for therapeutic applications in a range of diseases.
Collapse
Affiliation(s)
- Molly Klimak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
| | - Amanda Cimino
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
| | - Kristin L Lenz
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
| | - Luke E Springer
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, UT, 84108, USA
| | - Nathan Xu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA
| | - Christine T N Pham
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA.
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA.
- Center of Regenerative Medicine, Washington University in St. Louis, Campus Box 8233, Couch Biomedical Research Bldg., Room 3121, St. Louis, MO, 63110, USA.
| |
Collapse
|
6
|
Enayati M, Liu W, Madry H, Neisiany RE, Cucchiarini M. Functionalized hydrogels as smart gene delivery systems to treat musculoskeletal disorders. Adv Colloid Interface Sci 2024; 331:103232. [PMID: 38889626 DOI: 10.1016/j.cis.2024.103232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Despite critical advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy based on the delivery of therapeutic genetic sequences has strong value to offer effective, durable options to decisively manage such disorders. Furthermore, scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy, allowing for the spatiotemporal delivery of candidate genes to sites of injury. Among the many scaffolds for musculoskeletal research, hydrogels raised increasing attention in addition to other potent systems (solid, hybrid scaffolds) due to their versatility and competence as drug and cell carriers in tissue engineering and wound dressing. Attractive functionalities of hydrogels for musculoskeletal therapy include their injectability, stimuli-responsiveness, self-healing, and nanocomposition that may further allow to upgrade of them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. Such functionalized hydrogels may also be tuned to successfully transfer therapeutic genes in a minimally invasive manner in order to protect their cargos and allow for their long-term effects. In light of such features, this review focuses on functionalized hydrogels and demonstrates their competence for the treatment of musculoskeletal disorders using gene therapy procedures, from gene therapy principles to hydrogel functionalization methods and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are being discussed in the perspective of translation in patients. STATEMENT OF SIGNIFICANCE: Despite advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy has strong value in offering effective, durable options to decisively manage such disorders. Scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy. Among many scaffolds for musculoskeletal research, hydrogels raised increasing attention. Functionalities including injectability, stimuli-responsiveness, and self-healing, tune them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. This review introduces functionalized hydrogels for musculoskeletal disorder treatment using gene therapy procedures, from gene therapy principles to functionalized hydrogels and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are discussed from the perspective of translation in patients.
Collapse
Affiliation(s)
- Mohammadsaeid Enayati
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Rasoul Esmaeely Neisiany
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland; Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany.
| |
Collapse
|
7
|
Carneiro DC, Rocha VPC, Damasceno PKF, Barbosa JDV, Soares MBP. Therapeutic applications of synthetic gene/genetic circuits: a patent review. Front Bioeng Biotechnol 2024; 12:1425529. [PMID: 39161351 PMCID: PMC11330796 DOI: 10.3389/fbioe.2024.1425529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
A significant limitation of numerous current genetic engineering therapy approaches is their limited control over the strength, timing, or cellular context of their therapeutic effect. Synthetic gene/genetic circuits are synthetic biology approaches that can control the generation, transformation, or depletion of a specific DNA, RNA, or protein and provide precise control over gene expression and cellular behavior. They can be designed to perform logical operations by carefully selecting promoters, repressors, and other genetic components. Patent search was performed in Espacenet, resulting in 38 selected patents with 15 most frequent international classifications. Patent embodiments were categorized into applications for the delivery of therapeutic molecules, treatment of infectious diseases, treatment of cancer, treatment of bleeding, and treatment of metabolic disorders. The logic gates of selected genetic circuits are described to comprehensively demonstrate their therapeutic applications. Synthetic gene circuits can be customized for precise control of therapeutic interventions, leading to personalized therapies that respond specifically to individual patient needs, enhancing treatment efficacy and minimizing side effects. They can be highly sensitive biosensors that provide real-time therapy by accurate monitoring various biomarkers or pathogens and appropriately synthesizing a therapeutic molecule. Synthetic gene circuits may also lead to the development of advanced regenerative therapies and to implantable biodevices that produce on-demand bioactive molecules. However, this technology faces challenges for commercial profitability. The genetic circuit designs need adjustments for specific applications, and may have disadvantages like toxicity from multiple regulators, homologous recombination, context dependency, resource overuse, and environmental variability.
Collapse
Affiliation(s)
| | - Vinícius P. C. Rocha
- SENAI Institute of Advanced Health Systems Innovation, University Center SENAI CIMATEC, Salvador, Brazil
| | - Patrícia K. F. Damasceno
- SENAI Institute of Advanced Health Systems Innovation, University Center SENAI CIMATEC, Salvador, Brazil
| | - Josiane D. V. Barbosa
- SENAI Institute of Advanced Health Systems Innovation, University Center SENAI CIMATEC, Salvador, Brazil
| | - Milena B. P. Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
- SENAI Institute of Advanced Health Systems Innovation, University Center SENAI CIMATEC, Salvador, Brazil
| |
Collapse
|
8
|
Klimak M, Cimino A, Lenz K, Springer L, Collins K, Harasymowicz N, Xu N, Pham C, Guilak F. Engineered Self-Regulating Macrophages for Targeted Anti-inflammatory Drug Delivery. RESEARCH SQUARE 2024:rs.3.rs-4385938. [PMID: 38854124 PMCID: PMC11160898 DOI: 10.21203/rs.3.rs-4385938/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by increased levels of inflammation that primarily manifests in the joints. Macrophages act as key drivers for the progression of RA, contributing to the perpetuation of chronic inflammation and dysregulation of pro-inflammatory cytokines such as interleukin 1 (IL-1). The goal of this study was to develop a macrophage-based cell therapy for biologic drug delivery in an autoregulated manner. Methods For proof-of-concept, we developed "smart" macrophages to mitigate the effects of IL-1 by delivering its inhibitor, IL-1 receptor antagonist (IL-1Ra). Bone marrow-derived macrophages were lentivirally transduced with a synthetic gene circuit that uses an NF-κB inducible promoter upstream of either the Il1rn or firefly luciferase transgenes. Two types of joint like cells were utilized to examine therapeutic protection in vitro, miPSCs derived cartilage and isolated primary mouse synovial fibroblasts while the K/BxN mouse model of RA was utilized to examine in vivo therapeutic protection. Results These engineered macrophages were able to repeatably produce therapeutic levels of IL-1Ra that could successfully mitigate inflammatory activation in co-culture with both tissue engineered cartilage constructs and synovial fibroblasts. Following injection in vivo, macrophages homed to sites of inflammation and mitigated disease severity in the K/BxN mouse model of RA. Conclusion These findings demonstrate the successful development of engineered macrophages that possess the ability for controlled, autoregulated production of IL-1 based on inflammatory signaling such as the NF-κB pathway to mitigate the effects of this cytokine for applications in RA or other inflammatory diseases. This system provides proof of concept for applications in other immune cell types as self-regulating delivery systems for therapeutic applications in a range of diseases.
Collapse
|
9
|
Grol MW. The evolving landscape of gene therapy strategies for the treatment of osteoarthritis. Osteoarthritis Cartilage 2024; 32:372-384. [PMID: 38199296 DOI: 10.1016/j.joca.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/05/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
OBJECTIVES Significant advances have been made in our understanding of osteoarthritis (OA) pathogenesis; however, no disease-modifying therapies have been identified. This review will summarize the gene therapy landscape, its initial successes for OA, and possible challenges using recent studies and examples of gene therapies in clinical trials. DESIGN This narrative review has three major sections: 1) vector systems for OA gene therapy, 2) current and emerging targets for OA gene therapy, and 3) considerations and future directions. RESULTS Gene therapy is the strategy by which nucleic acids are delivered to treat and reverse disease progression. Specificity and prolonged expression of these nucleic acids are achieved by manipulating promoters, genes, and vector systems. Certain vector systems also allow for the development of combinatorial nucleic acid strategies that can be delivered in a single intraarticular injection - an approach likely required to treat the complexity of OA pathogenesis. Several viral and non-viral vector-based gene therapies are in clinical trials for OA, and many more are being evaluated in the preclinical arena. CONCLUSIONS In a post-coronavirus disease 2019 (COVID-19) era, the future of gene therapy for OA is certainly promising; however, the majority of preclinical validation continues to focus heavily on post-traumatic models and changes in only cartilage and subchondral bone. To ensure successful translation, new candidates in the preclinical arena should be examined against all joint tissues as well as pain using diverse models of injury-, obesity-, and age-induced disease. Lastly, consideration must be given to strategies for repeat administration and the cost of treatment owing to the chronic nature of OA.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
10
|
Walton BL, Shattuck-Brandt R, Hamann CA, Tung VW, Colazo JM, Brand DD, Hasty KA, Duvall CL, Brunger JM. A programmable arthritis-specific receptor for guided articular cartilage regenerative medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578281. [PMID: 38352576 PMCID: PMC10862827 DOI: 10.1101/2024.01.31.578281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Objective Investigational cell therapies have been developed as disease-modifying agents for the treatment of osteoarthritis (OA), including those that inducibly respond to inflammatory factors driving OA progression. However, dysregulated inflammatory cascades do not specifically signify the presence of OA. Here, we deploy a synthetic receptor platform that regulates cell behaviors in an arthritis-specific fashion to confine transgene expression to sites characterized by cartilage degeneration. Methods An scFv specific for type II collagen (CII) was used to produce a synthetic Notch (synNotch) receptor that enables "CII-synNotch" mesenchymal stromal cells (MSCs) to recognize CII fibers exposed in damaged cartilage. Engineered cell activation by both CII-treated culture surfaces and on primary tissue samples was measured via inducible reporter transgene expression. TGFβ3-expressing cells were assessed for cartilage anabolic gene expression via qRT-PCR. In a co-culture with CII-synNotch MSCs engineered to express IL-1Ra, ATDC5 chondrocytes were stimulated with IL-1α, and inflammatory responses of ATDC5s were profiled via qRT-PCR and an NF-κB reporter assay. Results CII-synNotch MSCs are highly responsive to CII, displaying activation ranges over 40-fold in response to physiologic CII inputs. CII-synNotch cells exhibit the capacity to distinguish between healthy and damaged cartilage tissue and constrain transgene expression to regions of exposed CII fibers. Receptor-regulated TGFβ3 expression resulted in upregulation of Acan and Col2a1 in MSCs, and inducible IL-1Ra expression by engineered CII-synNotch MSCs reduced pro-inflammatory gene expression in chondrocytes. Conclusion This work demonstrates proof-of-concept that the synNotch platform guides MSCs for spatially regulated, disease-dependent delivery of OA-relevant biologic drugs.
Collapse
Affiliation(s)
- Bonnie L. Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | | | - Catherine A. Hamann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Victoria W. Tung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - David D. Brand
- Research Service, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Karen A. Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis VA Medical Center, Memphis, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA
| |
Collapse
|
11
|
DeJulius CR, Walton BL, Colazo JM, d'Arcy R, Francini N, Brunger JM, Duvall CL. Engineering approaches for RNA-based and cell-based osteoarthritis therapies. Nat Rev Rheumatol 2024; 20:81-100. [PMID: 38253889 PMCID: PMC11129836 DOI: 10.1038/s41584-023-01067-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 01/24/2024]
Abstract
Osteoarthritis (OA) is a chronic, debilitating disease that substantially impairs the quality of life of affected individuals. The underlying mechanisms of OA are diverse and are becoming increasingly understood at the systemic, tissue, cellular and gene levels. However, the pharmacological therapies available remain limited, owing to drug delivery barriers, and consist mainly of broadly immunosuppressive regimens, such as corticosteroids, that provide only short-term palliative benefits and do not alter disease progression. Engineered RNA-based and cell-based therapies developed with synthetic chemistry and biology tools provide promise for future OA treatments with durable, efficacious mechanisms of action that can specifically target the underlying drivers of pathology. This Review highlights emerging classes of RNA-based technologies that hold potential for OA therapies, including small interfering RNA for gene silencing, microRNA and anti-microRNA for multi-gene regulation, mRNA for gene supplementation, and RNA-guided gene-editing platforms such as CRISPR-Cas9. Various cell-engineering strategies are also examined that potentiate disease-dependent, spatiotemporally regulated production of therapeutic molecules, and a conceptual framework is presented for their application as OA treatments. In summary, this Review highlights modern genetic medicines that have been clinically approved for other diseases, in addition to emerging genome and cellular engineering approaches, with the goal of emphasizing their potential as transformative OA treatments.
Collapse
Affiliation(s)
- Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Richard d'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
12
|
Ely EV, Kapinski AT, Paradi SG, Tang R, Guilak F, Collins KH. Designer Fat Cells: Adipogenic Differentiation of CRISPR-Cas9 Genome-Engineered Induced Pluripotent Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564206. [PMID: 37961399 PMCID: PMC10634849 DOI: 10.1101/2023.10.26.564206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Adipose tissue is an active endocrine organ that can signal bidirectionally to many tissues and organ systems in the body. With obesity, adipose tissue is a source of low-level inflammation that contributes to various co-morbidities and damage to downstream effector tissues. The ability to synthesize genetically engineered adipose tissue could have critical applications in studying adipokine signaling and the use of adipose tissue for novel therapeutic strategies. This study aimed to develop a method for non-viral adipogenic differentiation of genome-edited murine induced pluripotent stem cells (iPSCs) and to test the ability of such cells to engraft in mice in vivo . Designer adipocytes were created from iPSCs, which can be readily genetically engineered using CRISPR-Cas9 to knock out or insert individual genes of interest. As a model system for adipocyte-based drug delivery, an existing iPSC cell line that transcribes interleukin 1 receptor antagonist under the endogenous macrophage chemoattractant protein-1 promoter was tested for adipogenic capabilities under these same differentiation conditions. To understand the role of various adipocyte subtypes and their impact on health and disease, an efficient method was devised for inducing browning and whitening of IPSC-derived adipocytes in culture. Finally, to study the downstream effects of designer adipocytes in vivo , we transplanted the designer adipocytes into fat-free lipodystrophic mice as a model system for studying adipose signaling in different models of disease or repair. This novel translational tissue engineering and regenerative medicine platform provides an innovative approach to studying the role of adipose interorgan communication in various conditions.
Collapse
|
13
|
Ely E, Kapinski A, Paradi S, Tang R, Guilak F, Collins K. DESIGNER FAT CELLS: ADIPOGENIC DIFFERENTIATION OF CRISPR-CAS9 GENOME-ENGINEERED INDUCED PLURIPOTENT STEM CELLS. Eur Cell Mater 2023; 46:171-194. [PMID: 39830040 PMCID: PMC11741189 DOI: 10.22203/ecm.v046a09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Adipose tissue is an active endocrine organ that can signal bidirectionally to many tissues and organ systems in the body. With obesity, adipose tissue can serve as a source of low-level inflammation that contributes to various co-morbidities and damage to downstream effector tissues. The ability to synthesize genetically engineered adipose tissue could have critical applications in studying adipokine signaling and the use of adipose tissue for novel therapeutic strategies. This study aimed to develop a method for non-viral adipogenic differentiation of genome-edited murine induced pluripotent stem cells (iPSCs) and to test the ability of such cells to engraft in mice in vivo. Designer adipocytes were created from iPSCs, which can be readily genetically engineered using CRISPR-Cas9 to knock out or insert individual genes of interest. As a model system for adipocyte-based drug delivery, an existing iPSC cell line that transcribes interleukin 1 receptor antagonist under the endogenous macrophage chemoattractant protein-1 promoter was tested for adipogenic capabilities under these same differentiation conditions. To understand the role of various adipocyte subtypes and their impact on health and disease, an efficient method was devised for inducing browning and whitening of Ipsc-derived adipocytes in culture. Finally, to study the downstream effects of designer adipocytes in vivo, we transplanted the designer adipocytes into fat-free lipodystrophic mice as a model system for studying adipose signaling in different models of disease or repair. This novel translational tissue engineering and regenerative medicine platform provides an innovative approach to studying the role of adipose interorgan communication in various conditions.
Collapse
Affiliation(s)
- E.V. Ely
- Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| | - A.T. Kapinski
- Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| | - S.G. Paradi
- Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - R. Tang
- Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| | - F. Guilak
- Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
| | - K.H. Collins
- Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, MO 63110, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
14
|
Muthu S, Korpershoek JV, Novais EJ, Tawy GF, Hollander AP, Martin I. Failure of cartilage regeneration: emerging hypotheses and related therapeutic strategies. Nat Rev Rheumatol 2023; 19:403-416. [PMID: 37296196 DOI: 10.1038/s41584-023-00979-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/12/2023]
Abstract
Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.
Collapse
Affiliation(s)
- Sathish Muthu
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, New Delhi, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Jasmijn V Korpershoek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Emanuel J Novais
- Unidade Local de Saúde do Litoral Alentejano, Orthopedic Department, Santiago do Cacém, Portugal
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gwenllian F Tawy
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, UK
| | - Anthony P Hollander
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
15
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
16
|
Dicks AR, Steward N, Guilak F, Wu CL. Chondrogenic Differentiation of Human-Induced Pluripotent Stem Cells. Methods Mol Biol 2023; 2598:87-114. [PMID: 36355287 PMCID: PMC9830630 DOI: 10.1007/978-1-0716-2839-3_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The generation of large quantities of genetically defined human chondrocytes remains a critical step for the development of tissue engineering strategies for cartilage regeneration and high-throughput drug screening. This protocol describes chondrogenic differentiation of human-induced pluripotent stem cells (hiPSCs), which can undergo genetic modification and the capacity for extensive cell expansion. The hiPSCs are differentiated in a stepwise manner in monolayer through the mesodermal lineage for 12 days using defined growth factors and small molecules. This is followed by 28 days of chondrogenic differentiation in a 3D pellet culture system using transforming growth factor beta 3 and specific compounds to inhibit off-target differentiation. The 6-week protocol results in hiPSC-derived cartilaginous tissue that can be characterized by histology, immunohistochemistry, and gene expression or enzymatically digested to isolate chondrocyte-like cells. Investigators can use this protocol for experiments including genetic engineering, in vitro disease modeling, or tissue engineering.
Collapse
Affiliation(s)
- Amanda R Dicks
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
17
|
Geevarghese R, Sajjadi SS, Hudecki A, Sajjadi S, Jalal NR, Madrakian T, Ahmadi M, Włodarczyk-Biegun MK, Ghavami S, Likus W, Siemianowicz K, Łos MJ. Biodegradable and Non-Biodegradable Biomaterials and Their Effect on Cell Differentiation. Int J Mol Sci 2022; 23:ijms232416185. [PMID: 36555829 PMCID: PMC9785373 DOI: 10.3390/ijms232416185] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Biomaterials for tissue scaffolds are key components in modern tissue engineering and regenerative medicine. Targeted reconstructive therapies require a proper choice of biomaterial and an adequate choice of cells to be seeded on it. The introduction of stem cells, and the transdifferentiation procedures, into regenerative medicine opened a new era and created new challenges for modern biomaterials. They must not only fulfill the mechanical functions of a scaffold for implanted cells and represent the expected mechanical strength of the artificial tissue, but furthermore, they should also assure their survival and, if possible, affect their desired way of differentiation. This paper aims to review how modern biomaterials, including synthetic (i.e., polylactic acid, polyurethane, polyvinyl alcohol, polyethylene terephthalate, ceramics) and natural (i.e., silk fibroin, decellularized scaffolds), both non-biodegradable and biodegradable, could influence (tissue) stem cells fate, regulate and direct their differentiation into desired target somatic cells.
Collapse
Affiliation(s)
- Rency Geevarghese
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Seyedeh Sara Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | - Andrzej Hudecki
- Łukasiewicz Network-Institute of Non-Ferrous Metals, 44-121 Gliwice, Poland
| | - Samad Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | | | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Małgorzata K. Włodarczyk-Biegun
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Saeid Ghavami
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Wirginia Likus
- Department of Anatomy, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| | - Krzysztof Siemianowicz
- Department of Biochemistry, Faculty of Medicine in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| |
Collapse
|
18
|
Pferdehirt L, Guo P, Lu A, Huard M, Guilak F, Huard J. In vitro analysis of genome-engineered muscle-derived stem cells for autoregulated anti-inflammatory and antifibrotic activity. J Orthop Res 2022; 40:2937-2946. [PMID: 35293626 PMCID: PMC9477979 DOI: 10.1002/jor.25311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/19/2022] [Accepted: 02/21/2022] [Indexed: 02/04/2023]
Abstract
Traumatic muscle injury leads to chronic and pathologic fibrosis in skeletal muscles, primarily driven through upregulation of transforming growth factor-β1 (TGF-β1). Cell-based therapies, such as injection of muscle-derived stem cells (MDSCs), have shown promise in muscle repair. However, injected MDSCs in injured skeletal muscle can differentiate into myofibroblasts under the influence of TGF-β1, and contribute to the development of fibrosis, limiting their regenerative potential. In this study, we created a "smart" cell-based drug delivery system using CRISPR-Cas9 to genetically engineer MDSCs capable of sensing TGF-β1 and producing an antifibrotic biologic, decorin. These gene-edited smart cells, capable of inhibiting fibrosis in a dose-dependent and autoregulating manner, show anti-inflammatory and antifibrotic properties in vitro, without changing the expression of myogenic and stem cell markers as well as their cell proliferation and myogenic differentiation. Additionally, differentiation down a fibrotic lineage is reduced or eliminated in response to TGF-β1. Our results show that gene editing can be used to successfully create smart stem cells capable of producing biologic drugs with antifibrotic capabilities in a controlled and localized manner. This system provides a tool for cell-based drug delivery as the basis for a novel therapeutic approach for a variety of diseases.
Collapse
Affiliation(s)
- Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University,
St. Louis, MO 63110, USA
- Shriners Hospitals for Children – St. Louis, St.
Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington
University, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University, St.
Louis, MO 63110, USA
| | - Ping Guo
- Center for Regenerative & Personalized Medicine,
Steadman Philippon Research Institute; Vail, CO, 81657, USA
| | - Aiping Lu
- Center for Regenerative & Personalized Medicine,
Steadman Philippon Research Institute; Vail, CO, 81657, USA
| | - Mathew Huard
- Center for Regenerative & Personalized Medicine,
Steadman Philippon Research Institute; Vail, CO, 81657, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University,
St. Louis, MO 63110, USA
- Shriners Hospitals for Children – St. Louis, St.
Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington
University, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University, St.
Louis, MO 63110, USA
| | - Johnny Huard
- Center for Regenerative & Personalized Medicine,
Steadman Philippon Research Institute; Vail, CO, 81657, USA
| |
Collapse
|
19
|
Pferdehirt L, Damato AR, Dudek M, Meng QJ, Herzog ED, Guilak F. Synthetic gene circuits for preventing disruption of the circadian clock due to interleukin-1-induced inflammation. SCIENCE ADVANCES 2022; 8:eabj8892. [PMID: 35613259 PMCID: PMC9132444 DOI: 10.1126/sciadv.abj8892] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 04/07/2022] [Indexed: 05/16/2023]
Abstract
The circadian clock regulates tissue homeostasis through temporal control of tissue-specific clock-controlled genes. In articular cartilage, disruptions in the circadian clock are linked to a procatabolic state. In the presence of inflammation, the cartilage circadian clock is disrupted, which further contributes to the pathogenesis of diseases such as osteoarthritis. Using synthetic biology and tissue engineering, we developed and tested genetically engineered cartilage from murine induced pluripotent stem cells (miPSCs) capable of preserving the circadian clock in the presence of inflammation. We found that circadian rhythms arise following chondrogenic differentiation of miPSCs. Exposure of tissue-engineered cartilage to the inflammatory cytokine interleukin-1 (IL-1) disrupted circadian rhythms and degraded the cartilage matrix. All three inflammation-resistant approaches showed protection against IL-1-induced degradation and loss of circadian rhythms. These synthetic gene circuits reveal a unique approach to support daily rhythms in cartilage and provide a strategy for creating cell-based therapies to preserve the circadian clock.
Collapse
Affiliation(s)
- Lara Pferdehirt
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| | - Anna R. Damato
- Department of Biology, Washington University, St. Louis, MO 63130, USA
| | - Michal Dudek
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Erik D. Herzog
- Department of Biology, Washington University, St. Louis, MO 63130, USA
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children–St. Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
20
|
Engineering Closed-Loop, Autoregulatory Gene Circuits for Osteoarthritis Cell-Based Therapies. Curr Rheumatol Rep 2022; 24:96-110. [PMID: 35404006 DOI: 10.1007/s11926-022-01061-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Genetic engineering offers the possibility to simultaneously target multiple cellular pathways in the joints affected by osteoarthritis (OA). The purpose of this review is to summarize the ongoing efforts to develop disease-modifying osteoarthritis drugs (DMOADs) using genetic engineering, including targeting approaches, genome editing techniques, and delivery methods. RECENT FINDINGS Several gene circuits have been developed that reprogram cells to autonomously target inflammation, and their efficacy has been demonstrated in chondrocytes and stem cells. Gene circuits developed for metabolic disorders, such as those targeting insulin resistance and obesity, also have the potential to mitigate the impact of these conditions on OA onset and/or progression. Despite the strides made in characterizing the inflammatory environment of the OA joint, our incomplete understanding of how the multiple regulators interact to control signal transduction, gene transcription, and translation to protein limits the development of targeted disease-modifying therapeutics. Continuous advances in targeted genome editing, combined with online toolkits that simplify the design and production of gene circuits, have the potential to accelerate the discovery and clinical application of multi-target gene circuits with disease-modifying properties for the treatment of OA.
Collapse
|
21
|
Nims RJ, Pferdehirt L, Guilak F. Mechanogenetics: harnessing mechanobiology for cellular engineering. Curr Opin Biotechnol 2022; 73:374-379. [PMID: 34735987 PMCID: PMC10061441 DOI: 10.1016/j.copbio.2021.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 01/28/2023]
Abstract
'Mechanogenetics,' a new field at the convergence of mechanobiology and synthetic biology, presents an innovative strategy to treat, repair, or restore diseased cells and tissues by harnessing mechanical signal transduction pathways to control gene expression. While the role of mechanical forces in regulating development, homeostasis, and disease is well established, only recently have we identified the specific mechanosensors and downstream signaling pathways involved in these processes. Simultaneously, synthetic biological systems are developing increasingly sophisticated approaches of controlling mammalian cellular responses. Continued mechanistic refinement and identification of how cellular mechanosensors respond to homeostatic and pathological mechanical forces, combined with synthetic tools to integrate and respond to these inputs, promises to extend the development of new therapeutic approaches for treating disease.
Collapse
Affiliation(s)
- Robert J Nims
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Shriners Hospitals for Children - Saint Louis, St. Louis, MO, 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lara Pferdehirt
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Shriners Hospitals for Children - Saint Louis, St. Louis, MO, 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, 63105, USA
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Shriners Hospitals for Children - Saint Louis, St. Louis, MO, 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, 63105, USA.
| |
Collapse
|
22
|
Klimak M, Nims RJ, Pferdehirt L, Collins KH, Harasymowicz NS, Oswald SJ, Setton LA, Guilak F. Immunoengineering the next generation of arthritis therapies. Acta Biomater 2021; 133:74-86. [PMID: 33823324 DOI: 10.1016/j.actbio.2021.03.062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022]
Abstract
Immunoengineering continues to revolutionize healthcare, generating new approaches for treating previously intractable diseases, particularly in regard to cancer immunotherapy. In joint diseases, such as osteoarthritis (OA) and rheumatoid arthritis (RA), biomaterials and anti-cytokine treatments have previously been at that forefront of therapeutic innovation. However, while many of the existing anti-cytokine treatments are successful for a subset of patients, these treatments can also pose severe risks, adverse events and off-target effects due to continuous delivery at high dosages or a lack of disease-specific targets. The inadequacy of these current treatments has motivated the development of new immunoengineering strategies that offer safer and more efficacious alternative therapies through the precise and controlled targeting of specific upstream immune responses, including direct and mechanistically-driven immunoengineering approaches. Advances in the understanding of the immunomodulatory pathways involved in musculoskeletal disease, in combination with the growing emphasis on personalized medicine, stress the need for carefully considering the delivery strategies and therapeutic targets when designing therapeutics to better treat RA and OA. Here, we focus on recent advances in biomaterial and cell-based immunomodulation, in combination with genetic engineering, for therapeutic applications in joint diseases. The application of immunoengineering principles to the study of joint disease will not only help to elucidate the mechanisms of disease pathogenesis but will also generate novel disease-specific therapeutics by harnessing cellular and biomaterial responses. STATEMENT OF SIGNIFICANCE: It is now apparent that joint diseases such as osteoarthritis and rheumatoid arthritis involve the immune system at both local (i.e., within the joint) and systemic levels. In this regard, targeting the immune system using both biomaterial-based or cellular approaches may generate new joint-specific treatment strategies that are well-controlled, safe, and efficacious. In this review, we focus on recent advances in immunoengineering that leverage biomaterials and/or genetically engineered cells for therapeutic applications in joint diseases. The application of such approaches, especially synergistic strategies that target multiple immunoregulatory pathways, has the potential to revolutionize our understanding, treatment, and prevention of joint diseases.
Collapse
Affiliation(s)
- Molly Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Robert J Nims
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Sara J Oswald
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Lori A Setton
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
Choi YR, Collins KH, Springer LE, Pferdehirt L, Ross AK, Wu CL, Moutos FT, Harasymowicz NS, Brunger JM, Pham CTN, Guilak F. A genome-engineered bioartificial implant for autoregulated anticytokine drug delivery. SCIENCE ADVANCES 2021; 7:eabj1414. [PMID: 34516920 PMCID: PMC8442875 DOI: 10.1126/sciadv.abj1414] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/19/2021] [Indexed: 05/28/2023]
Abstract
Biologic drug therapies are increasingly used for inflammatory diseases such as rheumatoid arthritis but may cause significant adverse effects when delivered continuously at high doses. We used CRISPR-Cas9 genome editing of iPSCs to create a synthetic gene circuit that senses changing levels of endogenous inflammatory cytokines to trigger a proportional therapeutic response. Cells were engineered into cartilaginous constructs that showed rapid activation and recovery in response to inflammation in vitro or in vivo. In the murine K/BxN model of inflammatory arthritis, bioengineered implants significantly mitigated disease severity as measured by joint pain, structural damage, and systemic and local inflammation. Therapeutic implants completely prevented increased pain sensitivity and bone erosions, a feat not achievable by current clinically available disease-modifying drugs. Combination tissue engineering and synthetic biology promises a range of potential applications for treating chronic diseases via custom-designed cells that express therapeutic transgenes in response to dynamically changing biological signals.
Collapse
Affiliation(s)
- Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Luke E. Springer
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Alison K. Ross
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - Natalia S. Harasymowicz
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Cytex Therapeutics Inc., Durham, NC 27704, USA
| |
Collapse
|
24
|
Willard VP, Leddy HA, Palmer D, Wu CL, Liedtke W, Guilak F. Transient receptor potential vanilloid 4 as a regulator of induced pluripotent stem cell chondrogenesis. Stem Cells 2021; 39:1447-1456. [PMID: 34427363 DOI: 10.1002/stem.3440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a polymodal calcium-permeable cation channel that is highly expressed in cartilage and is sensitive to a variety of extracellular stimuli. The expression of this channel has been associated with the process of chondrogenesis in adult stem cells as well as several cell lines. Here, we used a chondrogenic reporter (Col2a1-GFP) in murine induced pluripotent stem cells (iPSCs) to examine the hypothesis that TRPV4 serves as both a marker and a regulator of chondrogenesis. Over 21 days of chondrogenesis, iPSCs showed significant increases in Trpv4 expression along with the standard chondrogenic gene markers Sox9, Acan, and Col2a1, particularly in the green fluorescent protein positive (GFP+) chondroprogenitor subpopulation. Increased gene expression for Trpv4 was also reflected by the presence of TRPV4 protein and functional Ca2+ signaling. Daily activation of TRPV4 using the specific agonist GSK1016790A resulted in significant increases in cartilaginous matrix production. An improved understanding of the role of TRPV4 in chondrogenesis may provide new insights into the development of new therapeutic approaches for diseases of cartilage, such as osteoarthritis, or channelopathies and hereditary disorders that affect cartilage during development. Harnessing the role of TRPV4 in chondrogenesis may also provide a novel approach for accelerating stem cell differentiation in functional tissue engineering of cartilage replacements for joint repair.
Collapse
Affiliation(s)
| | - Holly A Leddy
- Shared Materials Instrumentation Facility, Duke University, Durham, North Carolina, USA
| | - Daniel Palmer
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA.,Department of Orthopaedic Surgery and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA
| | | | - Farshid Guilak
- Cytex Therapeutics, Inc, Durham, North Carolina, USA.,Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Li Z, Huang Z, Bai L. Cell Interplay in Osteoarthritis. Front Cell Dev Biol 2021; 9:720477. [PMID: 34414194 PMCID: PMC8369508 DOI: 10.3389/fcell.2021.720477] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic disease and a significant health concern that needs to be urgently solved. OA affects the cartilage and entire joint tissues, including the subchondral bone, synovium, and infrapatellar fat pads. The physiological and pathological changes in these tissues affect the occurrence and development of OA. Understanding complex crosstalk among different joint tissues and their roles in OA initiation and progression is critical in elucidating the pathogenic mechanism of OA. In this review, we begin with an overview of the role of chondrocytes, synovial cells (synovial fibroblasts and macrophages), mast cells, osteoblasts, osteoclasts, various stem cells, and engineered cells (induced pluripotent stem cells) in OA pathogenesis. Then, we discuss the various mechanisms by which these cells communicate, including paracrine signaling, local microenvironment, co-culture, extracellular vesicles (exosomes), and cell tissue engineering. We particularly focus on the therapeutic potential and clinical applications of stem cell-derived extracellular vesicles, which serve as modulators of cell-to-cell communication, in the field of regenerative medicine, such as cartilage repair. Finally, the challenges and limitations related to exosome-based treatment for OA are discussed. This article provides a comprehensive summary of key cells that might be targets of future therapies for OA.
Collapse
Affiliation(s)
- Zihao Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyu Huang
- Foreign Languages College, Shanghai Normal University, Shanghai, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Song S, He X, Wang J, Song H, Wang Y, Liu Y, Zhou Z, Yu Z, Miao D, Xue Y. A novel long noncoding RNA, TMEM92-AS1, promotes gastric cancer progression by binding to YBX1 to mediate CCL5. Mol Oncol 2021; 15:1256-1273. [PMID: 33247987 PMCID: PMC8024739 DOI: 10.1002/1878-0261.12863] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/04/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
Numerous studies have revealed that long noncoding RNAs (lncRNAs) with oncogene properties play vital roles in gastric cancer (GC). In this study, we aimed to elucidate the function of TMEM92-AS1 in GC progression and to investigate its underlying mechanisms. TMEM92-AS1 was filtered from the Gene Expression Omnibus database. GC tissues and adjacent normal tissues were used to detect the expression level of TMEM92-AS1. MTT, colony-formation assays, Edu, cell cycle, apoptosis and subcutaneous tumour formation assays were used to detect the role of TMEM92-AS1 in cell function. RNA transcriptome sequencing was used to seek downstream target genes. Reverse transcription (RT)-qPCR, western blot, RNA and chromatin immunoprecipitation assays were used to investigate the mechanisms involved. TMEM92-AS1 was significantly overexpressed in GC tissues and correlated with poor overall survival and disease-free survival. Furthermore, TMEM92-AS1 promoted GC cell proliferation and migration in vitro and tumorigenic ability in vivo. RNA transcriptome sequence analysis revealed a potential downstream target gene, C-C motif chemokine ligand 5 (CCL5), and a mechanistic study found that TMEM92-AS1 regulated CCL5 by binding to the transcription factor Y-box binding protein 1(YBX1), which has oncogene properties. In addition, TMEM92-AS1 was found to be associated with peripheral blood leukocyte counts, especially neutrophils. Further investigation found that TMEM92-AS1 may affect leukocytes via regulation of the expression of granulocyte colony-stimulating factor in GC tissues. Our data provide an in-depth insight into the mechanism behind the lncRNA TMEM92-AS1, how it promotes GC progression and the possible mechanism in affecting peripheral leukocyte counts. Therefore, TMEM92-AS1 is a potential target for GC individualized therapy and prognostic assessment.
Collapse
Affiliation(s)
- Shubin Song
- Department of Gastrointestinal SurgeryHarbin Medical University Cancer HospitalChina
- Department of Breast SurgeryShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Xuezhi He
- Department of Anatomy, Histology and EmbryologyThe Research Centre for Bone and Stem CellsNanjing Medical UniversityChina
| | - Jing Wang
- Department of Anatomy, Histology and EmbryologyState Key Laboratory of Reproductive MedicineThe Research Centre for Bone and Stem CellsNanjing Medical UniversityChina
| | - Hongtao Song
- Department of PathologyHarbin Medical University Cancer HospitalChina
| | - Yimin Wang
- Department of Gastrointestinal SurgeryHarbin Medical University Cancer HospitalChina
| | - Yansong Liu
- Department of Breast SurgeryShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Zhengbo Zhou
- Department of Breast SurgeryShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Zhiyong Yu
- Department of Breast SurgeryShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Dengshun Miao
- The Research Centre for AgeingFriendship Affiliated Plastic Surgery Hospital of Nanjing Medical UniversityChina
| | - Yingwei Xue
- Department of Gastrointestinal SurgeryHarbin Medical University Cancer HospitalChina
| |
Collapse
|
27
|
Nims RJ, Pferdehirt L, Ho NB, Savadipour A, Lorentz J, Sohi S, Kassab J, Ross AK, O'Conor CJ, Liedtke WB, Zhang B, McNulty AL, Guilak F. A synthetic mechanogenetic gene circuit for autonomous drug delivery in engineered tissues. SCIENCE ADVANCES 2021; 7:eabd9858. [PMID: 33571125 PMCID: PMC7840132 DOI: 10.1126/sciadv.abd9858] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/08/2020] [Indexed: 05/12/2023]
Abstract
Mechanobiologic signals regulate cellular responses under physiologic and pathologic conditions. Using synthetic biology and tissue engineering, we developed a mechanically responsive bioartificial tissue that responds to mechanical loading to produce a preprogrammed therapeutic biologic drug. By deconstructing the signaling networks induced by activation of the mechanically sensitive ion channel transient receptor potential vanilloid 4 (TRPV4), we created synthetic TRPV4-responsive genetic circuits in chondrocytes. We engineered these cells into living tissues that respond to mechanical loading by producing the anti-inflammatory biologic drug interleukin-1 receptor antagonist. Chondrocyte TRPV4 is activated by osmotic loading and not by direct cellular deformation, suggesting that tissue loading is transduced into an osmotic signal that activates TRPV4. Either osmotic or mechanical loading of tissues transduced with TRPV4-responsive circuits protected constructs from inflammatory degradation by interleukin-1α. This synthetic mechanobiology approach was used to develop a mechanogenetic system to enable long-term, autonomously regulated drug delivery driven by physiologically relevant loading.
Collapse
Affiliation(s)
- Robert J Nims
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lara Pferdehirt
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| | - Noelani B Ho
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Alireza Savadipour
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63105, USA
| | - Jeremiah Lorentz
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| | - Sima Sohi
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| | - Jordan Kassab
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| | - Alison K Ross
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| | - Christopher J O'Conor
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wolfgang B Liedtke
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amy L McNulty
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
28
|
Wang M, Luo Y, Yu Y, Chen F. Bioengineering Approaches to Accelerate Clinical Translation of Stem Cell Therapies Treating Osteochondral Diseases. Stem Cells Int 2020; 2020:8874742. [PMID: 33424981 PMCID: PMC7775142 DOI: 10.1155/2020/8874742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/17/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
The osteochondral tissue is an interface between articular cartilage and bone. The diverse composition, mechanical properties, and cell phenotype in these two tissues pose a big challenge for the reconstruction of the defected interface. Due to the availability and inherent regenerative therapeutic properties, stem cells provide tremendous promise to repair osteochondral defect. This review is aimed at highlighting recent progress in utilizing bioengineering approaches to improve stem cell therapies for osteochondral diseases, which include microgel encapsulation, adhesive bioinks, and bioprinting to control the administration and distribution. We will also explore utilizing synthetic biology tools to control the differentiation fate and deliver therapeutic biomolecules to modulate the immune response. Finally, future directions and opportunities in the development of more potent and predictable stem cell therapies for osteochondral repair are discussed.
Collapse
Affiliation(s)
- Meng Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yixuan Luo
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yin Yu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
29
|
|
30
|
Wu CL, Harasymowicz NS, Klimak MA, Collins KH, Guilak F. The role of macrophages in osteoarthritis and cartilage repair. Osteoarthritis Cartilage 2020; 28:544-554. [PMID: 31926267 PMCID: PMC7214213 DOI: 10.1016/j.joca.2019.12.007] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/15/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is a family of degenerative diseases affecting multiple joint tissues. Despite the diverse etiology and pathogenesis of OA, increasing evidence suggests that macrophages can play a significant role in modulating joint inflammation, and thus OA severity, via various secreted mediators. Recent advances in next-generation sequencing technologies coupled with proteomic and epigenetic tools have greatly facilitated research to elucidate the embryonic origin of macrophages in various tissues including joint synovium. Furthermore, scientists have now begun to appreciate that macrophage polarization can span beyond the conventionally recognized binary states (i.e., pro-inflammatory M1-like vs anti-inflammatory M2-like) and may encompass a broad spectrum of phenotypes. Although the presence of these cells has been shown in multiple joint tissues, additional mechanistic studies are required to provide a comprehensive understanding of the precise role of these diverse macrophage populations in OA onset and progression. New approaches that can modulate macrophages into desired functional phenotypes may provide novel therapeutic strategies for preventing OA or enhancing cartilage repair and regeneration.
Collapse
Affiliation(s)
- C-L Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - N S Harasymowicz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - M A Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA
| | - K H Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - F Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
31
|
Farhang N, Silverman L, Bowles RD. Improving Cell Therapy Survival and Anabolism in Harsh Musculoskeletal Disease Environments. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:348-366. [PMID: 32070243 DOI: 10.1089/ten.teb.2019.0324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cell therapies are an up and coming technology in orthopedic medicine that has the potential to provide regenerative treatments for musculoskeletal disease. Despite numerous cell therapies showing preclinical success for common musculoskeletal indications of disc degeneration and osteoarthritis, there have been mixed results when testing these therapies in humans during clinical trials. A theory behind the mixed success of these cell therapies is that the harsh microenvironments of the disc and knee they are entering inhibit their anabolism and survival. Therefore, there is much ongoing research looking into how to improve the survival and anabolism of cell therapies within these musculoskeletal disease environments. This includes research into improving cell function under specific microenvironmental conditions known to exist in the intervertebral disc (IVD) and knee environment such as hypoxia, low-nutrient conditions, hyperosmolarity, acidity, and inflammation. This research also includes improving differentiation of cells into desired native cell phenotypes to better enhance their survival and anabolism in the knee and IVD. This review highlights the effects of specific musculoskeletal microenvironmental challenges on cell therapies and what research is being done to overcome these challenges. Impact statement While there has been significant clinical interest in using cell therapies for musculoskeletal pathologies in the knee and intervertebral disc, cell therapy clinical trials have had mixed outcomes. The information presented in this review includes the environmental challenges (i.e., acidic pH, inflammation, hyperosmolarity, hypoxia, and low nutrition) that cell therapies experience in these pathological musculoskeletal environments. This review summarizes studies that describe various approaches to improving the therapeutic capability of cell therapies in these harsh environments. The result is an overview of what approaches can be targeted and/or combined to develop a more consistent cell therapy for musculoskeletal pathologies.
Collapse
Affiliation(s)
- Niloofar Farhang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | | | - Robby D Bowles
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
32
|
Synthetic biology for improving cell fate decisions and tissue engineering outcomes. Emerg Top Life Sci 2019; 3:631-643. [PMID: 33523179 DOI: 10.1042/etls20190091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
Synthetic biology is a relatively new field of science that combines aspects of biology and engineering to create novel tools for the construction of biological systems. Using tools within synthetic biology, stem cells can then be reprogrammed and differentiated into a specified cell type. Stem cells have already proven to be largely beneficial in many different therapies and have paved the way for tissue engineering and regenerative medicine. Although scientists have made great strides in tissue engineering, there still remain many questions to be answered in regard to regeneration. Presented here is an overview of synthetic biology, common tools built within synthetic biology, and the way these tools are being used in stem cells. Specifically, this review focuses on how synthetic biologists engineer genetic circuits to dynamically control gene expression while also introducing emerging topics such as genome engineering and synthetic transcription factors. The findings mentioned in this review show the diverse use of stem cells within synthetic biology and provide a foundation for future research in tissue engineering with the use of synthetic biology tools. Overall, the work done using synthetic biology in stem cells is in its early stages, however, this early work is leading to new approaches for repairing diseased and damaged tissues and organs, and further expanding the field of tissue engineering.
Collapse
|
33
|
Guilak F, Pferdehirt L, Ross AK, Choi YR, Collins KH, Nims RJ, Katz DB, Klimak M, Tabbaa S, Pham CT. Designer Stem Cells: Genome Engineering and the Next Generation of Cell-Based Therapies. J Orthop Res 2019; 37:1287-1293. [PMID: 30977548 PMCID: PMC6546536 DOI: 10.1002/jor.24304] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 02/04/2023]
Abstract
Stem cells provide tremendous promise for the development of new therapeutic approaches for musculoskeletal conditions. In addition to their multipotency, certain types of stem cells exhibit immunomodulatory effects that can mitigate inflammation and enhance tissue repair. However, the translation of stem cell therapies to clinical practice has proven difficult due to challenges in intradonor and interdonor variability, engraftment, variability in recipient microenvironment and patient indications, and limited therapeutic biological activity. In this regard, the success of stem cell-based therapies may benefit from cellular engineering approaches to enhance factors such as purification, homing and cell survival, trophic effects, or immunomodulatory signaling. By combining recent advances in gene editing, synthetic biology, and tissue engineering, the potential exists to create new classes of "designer" cells that have prescribed cell-surface molecules and receptors as well as synthetic gene circuits that provide for autoregulated drug delivery or enhanced tissue repair. Published by Wiley Periodicals, Inc. J Orthop Res 37:1287-1293, 2019.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110,Correspondence: Farshid Guilak, Ph.D. Center of Regenerative Medicine, Washington University, St. Louis, Campus Box 8233, McKinley Research Bldg, Room 3121, St. Louis, MO 63110-1624.
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Alison K. Ross
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Robert J. Nims
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Dakota B. Katz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Molly Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | | | - Christine T.N. Pham
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, MO, 63110
| |
Collapse
|