1
|
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
|
2
|
Graham JP, Castro JG, Werba LC, Fardone LC, Francis KP, Ramamurthi A, Layden M, McCarthy HO, Gonzalez-Fernandez T. Versatile Cell Penetrating Peptide for Multimodal CRISPR Gene Editing in Primary Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614499. [PMID: 39386541 PMCID: PMC11463527 DOI: 10.1101/2024.09.23.614499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
CRISPR gene editing offers unprecedented genomic and transcriptomic control for precise regulation of cell function and phenotype. However, delivering the necessary CRISPR components to therapeutically relevant cell types without cytotoxicity or unexpected side effects remains challenging. Viral vectors risk genomic integration and immunogenicity while non-viral delivery systems are challenging to adapt to different CRISPR cargos, and many are highly cytotoxic. The arginine-alanine-leucine-alanine (RALA) cell penetrating peptide is an amphiphilic peptide that self-assembles into nanoparticles through electrostatic interactions with negatively charged molecules before delivering them across the cell membrane. This system has been used to deliver DNAs, RNAs, and small anionic molecules to primary cells with lower cytotoxicity compared to alternative non-viral approaches. Given the low cytotoxicity, versatility, and competitive transfection rates of RALA, we aimed to establish this peptide as a new CRISPR delivery system in a wide range of molecular formats across different editing modalities. We report that RALA was able to effectively encapsulate and deliver CRISPR in DNA, RNA, and ribonucleic protein (RNP) formats to primary mesenchymal stem cells (MSCs). Comparisons between RALA and commercially available reagents revealed superior cell viability leading to higher numbers of transfected cells and the maintenance of cell proliferative capacity. We then used the RALA peptide for the knock-in and knock-out of reporter genes into the MSC genome as well as for the transcriptional activation of therapeutically relevant genes. In summary, we establish RALA as a powerful tool for safer and effective delivery of CRISPR machinery in multiple cargo formats for a wide range of gene editing strategies.
Collapse
Affiliation(s)
- Josh P. Graham
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | | | - Lisette C. Werba
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Luke C. Fardone
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | | | - Anand Ramamurthi
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Michael Layden
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University Belfast, Northern Ireland, United Kingdom
| | | |
Collapse
|
3
|
Cao L, Gao W, Yang H, Zeng R, Yin Z. Adipocyte enhancer binding protein 1 knockdown alleviates osteoarthritis through inhibiting NF-κB signaling pathway-mediated inflammation and extracellular matrix degradation. J Cell Commun Signal 2024; 18:e12022. [PMID: 38946719 PMCID: PMC11208125 DOI: 10.1002/ccs3.12022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 07/02/2024] Open
Abstract
Inflammation promotes the degradation of the extracellular matrix, which contributes to the development of osteoarthritis (OA). Adipocyte enhancer binding protein 1 (AEBP1) participates in multiple pathological processes related to inflammatory diseases. However, the role of AEBP1 in OA development is unknown. We found a higher AEBP1 expression in articular cartilage of OA patients (n = 20) compared to their normal controls (n = 10). Thus, we inferred that AEBP1 might affect OA progression. Then mice with destabilization of the medial meniscus (DMM) surgery and chondrocytes with IL-1β treatment (10 ng/mL) were used to mimic OA. The increased AEBP1 expression was observed in models of OA. AEBP1 knockdown in chondrocytes reversed IL-1β-induced inflammation and extracellular matrix degradation, which was mediated by the inactivation of NF-κB signaling pathway and the increased IκBα activity. Co-immunoprecipitation assay indicated the interaction between AEBP1 and IκBα. Importantly, IκBα knockdown depleted the protective role of AEBP1 knockdown in OA. Moreover, AEBP1 knockdown in mice with OA showed similar results to those in chondrocytes. Collectively, our findings suggest that AEBP1 knockdown alleviates the development of OA, providing a novel strategy for OA treatment.
Collapse
Affiliation(s)
- Le Cao
- Department of OrthopedicsThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Department of OrthopedicsFuyang Hospital of Anhui Medical UniversityFuyangAnhuiChina
| | - Weilu Gao
- Department of OrthopedicsThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Haitao Yang
- Department of OrthopedicsFuyang Hospital of Anhui Medical UniversityFuyangAnhuiChina
| | - Ran Zeng
- Department of Intensive Care UnitFuyang Hospital of Anhui Medical UniversityFuyangAnhuiChina
| | - Zongsheng Yin
- Department of OrthopedicsThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
4
|
Mamachan M, Sharun K, Banu SA, Muthu S, Pawde AM, Abualigah L, Maiti SK. Mesenchymal stem cells for cartilage regeneration: Insights into molecular mechanism and therapeutic strategies. Tissue Cell 2024; 88:102380. [PMID: 38615643 DOI: 10.1016/j.tice.2024.102380] [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: 01/11/2024] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
The use of mesenchymal stem cells (MSCs) in cartilage regeneration has gained significant attention in regenerative medicine. This paper reviews the molecular mechanisms underlying MSC-based cartilage regeneration and explores various therapeutic strategies to enhance the efficacy of MSCs in this context. MSCs exhibit multipotent capabilities and can differentiate into various cell lineages under specific microenvironmental cues. Chondrogenic differentiation, a complex process involving signaling pathways, transcription factors, and growth factors, plays a pivotal role in the successful regeneration of cartilage tissue. The chondrogenic differentiation of MSCs is tightly regulated by growth factors and signaling pathways such as TGF-β, BMP, Wnt/β-catenin, RhoA/ROCK, NOTCH, and IHH (Indian hedgehog). Understanding the intricate balance between these pathways is crucial for directing lineage-specific differentiation and preventing undesirable chondrocyte hypertrophy. Additionally, paracrine effects of MSCs, mediated by the secretion of bioactive factors, contribute significantly to immunomodulation, recruitment of endogenous stem cells, and maintenance of chondrocyte phenotype. Pre-treatment strategies utilized to potentiate MSCs, such as hypoxic conditions, low-intensity ultrasound, kartogenin treatment, and gene editing, are also discussed for their potential to enhance MSC survival, differentiation, and paracrine effects. In conclusion, this paper provides a comprehensive overview of the molecular mechanisms involved in MSC-based cartilage regeneration and outlines promising therapeutic strategies. The insights presented contribute to the ongoing efforts in optimizing MSC-based therapies for effective cartilage repair.
Collapse
Affiliation(s)
- Merlin Mamachan
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India; Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan.
| | - S Amitha Banu
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Sathish Muthu
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India; Orthopaedic Research Group, Coimbatore, Tamil Nadu, India; Department of Orthopaedics, Government Medical College, Kaur, Tamil Nadu, India
| | - Abhijit M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laith Abualigah
- Artificial Intelligence and Sensing Technologies (AIST) Research Center, University of Tabuk, Tabuk 71491, Saudi Arabia; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan; Computer Science Department, Al al-Bayt University, Mafraq 25113, Jordan; MEU Research Unit, Middle East University, Amman 11831, Jordan; Department of Electrical and Computer Engineering, Lebanese American University, Byblos 13-5053, Lebanon; Applied Science Research Center, Applied Science Private University, Amman 11931, Jordan; School of Engineering and Technology, Sunway University Malaysia, Petaling Jaya 27500, Malaysia
| | - Swapan Kumar Maiti
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
5
|
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
|
6
|
Jia S, Liang R, Chen J, Liao S, Lin J, Li W. Emerging technology has a brilliant future: the CRISPR-Cas system for senescence, inflammation, and cartilage repair in osteoarthritis. Cell Mol Biol Lett 2024; 29:64. [PMID: 38698311 PMCID: PMC11067114 DOI: 10.1186/s11658-024-00581-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Osteoarthritis (OA), known as one of the most common types of aseptic inflammation of the musculoskeletal system, is characterized by chronic pain and whole-joint lesions. With cellular and molecular changes including senescence, inflammatory alterations, and subsequent cartilage defects, OA eventually leads to a series of adverse outcomes such as pain and disability. CRISPR-Cas-related technology has been proposed and explored as a gene therapy, offering potential gene-editing tools that are in the spotlight. Considering the genetic and multigene regulatory mechanisms of OA, we systematically review current studies on CRISPR-Cas technology for improving OA in terms of senescence, inflammation, and cartilage damage and summarize various strategies for delivering CRISPR products, hoping to provide a new perspective for the treatment of OA by taking advantage of CRISPR technology.
Collapse
Affiliation(s)
- Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Rongji Liang
- Shantou University Medical College, Shantou, 515041, China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Shuai Liao
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Wei Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
7
|
Kaur G, Wu B, Murali S, Lanigan T, Coleman RM. A synthetic, closed-looped gene circuit for the autonomous regulation of RUNX2 activity during chondrogenesis. FASEB J 2024; 38:e23484. [PMID: 38407380 PMCID: PMC10981937 DOI: 10.1096/fj.202300348rr] [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: 02/24/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
The transcription factor RUNX2 is a key regulator of chondrocyte phenotype during development, making it an ideal target for prevention of undesirable chondrocyte maturation in cartilage tissue-engineering strategies. Here, we engineered an autoregulatory gene circuit (cisCXp-shRunx2) that negatively controls RUNX2 activity in chondrogenic cells via RNA interference initiated by a tunable synthetic Col10a1-like promoter (cisCXp). The cisCXp-shRunx2 gene circuit is designed based on the observation that induced RUNX2 silencing after early chondrogenesis enhances the accumulation of cartilaginous matrix in ATDC5 cells. We show that the cisCXp-shRunx2 initiates RNAi of RUNX2 in maturing chondrocytes in response to the increasing intracellular RUNX2 activity without interfering with early chondrogenesis. The induced loss of RUNX2 activity in turn negatively regulates the gene circuit itself. Moreover, the efficacy of RUNX2 suppression from cisCXp-shRunx2 can be controlled by modifying the sensitivity of cisCXp promoter. Finally, we show the efficacy of inhibiting RUNX2 in preventing matrix loss in human mesenchymal stem cell-derived (hMSC-derived) cartilage under conditions that induce chondrocyte hypertrophic differentiation, including inflammation. Overall, our results demonstrated that the negative modulation of RUNX2 activity with our autoregulatory gene circuit enhanced matrix synthesis and resisted ECM degradation by reprogrammed MSC-derived chondrocytes in response to the microenvironment of the degenerative joint.
Collapse
Affiliation(s)
- Gurcharan Kaur
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Biming Wu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sunjana Murali
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Lanigan
- Biomedical Research Vector Core, University of Michigan, Ann Arbor, MI, USA
| | - Rhima M. Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Klimak M, Guilak F. Genetically engineered macrophages derived from iPSCs for self-regulating delivery of anti-inflammatory biologic drugs. J Tissue Eng Regen Med 2024; 2024:6201728. [PMID: 38571695 PMCID: PMC10990417 DOI: 10.1155/2024/6201728] [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] [Indexed: 04/05/2024]
Abstract
In rheumatoid arthritis, dysregulated cytokine signaling has been implicated as a primary factor in chronic inflammation. Many antirheumatic and biological therapies are used to suppress joint inflammation, but despite these advances, effectiveness is not universal, and delivery is often at high doses, which can predispose patients to significant off-target effects. During chronic inflammation, the inappropriate regulation of signaling factors by macrophages accelerates progression of disease by driving an imbalance of inflammatory cytokines, making macrophages an ideal cellular target. To develop a macrophage-based therapy to treat chronic inflammation, we engineered a novel induced pluripotent stem cell (iPSC)-derived macrophage capable of delivering soluble TNF receptor 1 (TNFR1), an anti-inflammatory biologic inhibitor of tumor necrosis factor alpha (TNF-α), in an auto-regulated manner in response to TNF-α. Murine iPSCs were differentiated into macrophages (iMACs) over a 17-day optimized protocol with continued successful differentiation confirmed at key timepoints. Varying inflammatory and immunomodulatory stimuli demonstrated traditional macrophage function and phenotypes. In response to TNF-α, therapeutic iMACs produced high levels of sTNFR1 in an autoregulated manner, which inhibited inflammatory signaling. This self-regulating iMAC system demonstrated the potential for macrophage-based drug delivery as a novel therapeutic approach for a variety of chronic inflammatory 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
| | - 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
|
9
|
Lotfi M, Morshedi Rad D, Mashhadi SS, Ashouri A, Mojarrad M, Mozaffari-Jovin S, Farrokhi S, Hashemi M, Lotfi M, Ebrahimi Warkiani M, Abbaszadegan MR. Recent Advances in CRISPR/Cas9 Delivery Approaches for Therapeutic Gene Editing of Stem Cells. Stem Cell Rev Rep 2023; 19:2576-2596. [PMID: 37723364 PMCID: PMC10661828 DOI: 10.1007/s12015-023-10585-3] [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] [Accepted: 06/30/2023] [Indexed: 09/20/2023]
Abstract
Rapid advancement in genome editing technologies has provided new promises for treating neoplasia, cardiovascular, neurodegenerative, and monogenic disorders. Recently, the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system has emerged as a powerful gene editing tool offering advantages, including high editing efficiency and low cost over the conventional approaches. Human pluripotent stem cells (hPSCs), with their great proliferation and differentiation potential into different cell types, have been exploited in stem cell-based therapy. The potential of hPSCs and the capabilities of CRISPR/Cas9 genome editing has been paradigm-shifting in medical genetics for over two decades. Since hPSCs are categorized as hard-to-transfect cells, there is a critical demand to develop an appropriate and effective approach for CRISPR/Cas9 delivery into these cells. This review focuses on various strategies for CRISPR/Cas9 delivery in stem cells.
Collapse
Affiliation(s)
- Malihe Lotfi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dorsa Morshedi Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - Samaneh Sharif Mashhadi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Ashouri
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Mozaffari-Jovin
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Farrokhi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Lotfi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia.
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, Australia.
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
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
|
11
|
Bonato A, Fisch P, Ponta S, Fercher D, Manninen M, Weber D, Eklund KK, Barreto G, Zenobi‐Wong M. Engineering Inflammation-Resistant Cartilage: Bridging Gene Therapy and Tissue Engineering. Adv Healthc Mater 2023; 12:e2202271. [PMID: 36841937 PMCID: PMC11468558 DOI: 10.1002/adhm.202202271] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 01/09/2023] [Indexed: 02/27/2023]
Abstract
Articular cartilage defects caused by traumatic injury rarely heal spontaneously and predispose into post-traumatic osteoarthritis. In the current autologous cell-based treatments the regenerative process is often hampered by the poor regenerative capacity of adult cells and the inflammatory state of the injured joint. The lack of ideal treatment options for cartilage injuries motivated the authors to tissue engineer a cartilage tissue which would be more resistant to inflammation. A clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 knockout of TGF-β-activated kinase 1 (TAK1) gene in polydactyly chondrocytes provides multivalent protection against the signals that activate the pro-inflammatory and catabolic NF-κB pathway. The TAK1-KO chondrocytes encapsulate into a hyaluronan hydrogel deposit copious cartilage extracellular matrix proteins and facilitate integration onto native cartilage, even under proinflammatory conditions. Furthermore, when implanted in vivo, compared to WT fewer pro-inflammatory M1 macrophages invade the cartilage, likely due to the lower levels of cytokines secreted by the TAK1-KO polydactyly chondrocytes. The engineered cartilage thus represents a new paradigm-shift for the creation of more potent and functional tissues for use in regenerative medicine.
Collapse
Affiliation(s)
- Angela Bonato
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | - Philipp Fisch
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | - Simone Ponta
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | - David Fercher
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | | | - Daniel Weber
- Division of Hand SurgeryUniversity Children's HospitalZürich8032Switzerland
| | - Kari K. Eklund
- Orton Orthopedic Hospital HelsinkiHelsinki00280Finland
- Department of RheumatologyUniversity of Helsinki and Helsinki University HospitalHelsinki00014Finland
| | - Goncalo Barreto
- Orton Orthopedic Hospital HelsinkiHelsinki00280Finland
- Translational Immunology Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinki00014Finland
| | - Marcy Zenobi‐Wong
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| |
Collapse
|
12
|
Collins KH, Pferdehirt L, Saleh LS, Savadipour A, Springer LE, Lenz KL, Thompson DM, Oswald SJ, Pham CTN, Guilak F. Hydrogel Encapsulation of Genome-Engineered Stem Cells for Long-Term Self-Regulating Anti-Cytokine Therapy. Gels 2023; 9:169. [PMID: 36826339 PMCID: PMC9956980 DOI: 10.3390/gels9020169] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Biologic therapies have revolutionized treatment options for rheumatoid arthritis (RA) but their continuous administration at high doses may lead to adverse events. Thus, the development of improved drug delivery systems that can sense and respond commensurately to disease flares represents an unmet medical need. Toward this end, we generated induced pluripotent stem cells (iPSCs) that express interleukin-1 receptor antagonist (IL-1Ra, an inhibitor of IL-1) in a feedback-controlled manner driven by the macrophage chemoattractant protein-1 (Ccl2) promoter. Cells were seeded in agarose hydrogel constructs made from 3D printed molds that can be injected subcutaneously via a blunt needle, thus simplifying implantation of the constructs, and the translational potential. We demonstrated that the subcutaneously injected agarose hydrogels containing genome-edited Ccl2-IL1Ra iPSCs showed significant therapeutic efficacy in the K/BxN model of inflammatory arthritis, with nearly complete abolishment of disease severity in the front paws. These implants also exhibited improved implant longevity as compared to the previous studies using 3D woven scaffolds, which require surgical implantation. This minimally invasive cell-based drug delivery strategy may be adapted for the treatment of other autoimmune or chronic diseases, potentially accelerating translation to the clinic.
Collapse
Affiliation(s)
- Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, 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, 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
| | - Leila S. Saleh
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63110, USA
| | - Luke E. Springer
- Division of Rheumatology, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Kristin L. Lenz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
| | - Dominic M. Thompson
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, 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, MO 63110, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Department of 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, 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
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
13
|
Amini M, Venkatesan JK, Liu W, Leroux A, Nguyen TN, Madry H, Migonney V, Cucchiarini M. Advanced Gene Therapy Strategies for the Repair of ACL Injuries. Int J Mol Sci 2022; 23:ijms232214467. [PMID: 36430947 PMCID: PMC9695211 DOI: 10.3390/ijms232214467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
The anterior cruciate ligament (ACL), the principal ligament for stabilization of the knee, is highly predisposed to injury in the human population. As a result of its poor intrinsic healing capacities, surgical intervention is generally necessary to repair ACL lesions, yet the outcomes are never fully satisfactory in terms of long-lasting, complete, and safe repair. Gene therapy, based on the transfer of therapeutic genetic sequences via a gene vector, is a potent tool to durably and adeptly enhance the processes of ACL repair and has been reported for its workability in various experimental models relevant to ACL injuries in vitro, in situ, and in vivo. As critical hurdles to the effective and safe translation of gene therapy for clinical applications still remain, including physiological barriers and host immune responses, biomaterial-guided gene therapy inspired by drug delivery systems has been further developed to protect and improve the classical procedures of gene transfer in the future treatment of ACL injuries in patients, as critically presented here.
Collapse
Affiliation(s)
- Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Amélie Leroux
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Tuan Ngoc Nguyen
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Véronique Migonney
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
- Correspondence: or
| |
Collapse
|
14
|
Nelson AL, Fontana G, Miclau E, Rongstad M, Murphy W, Huard J, Ehrhart N, Bahney C. Therapeutic approaches to activate the canonical Wnt pathway for bone regeneration. J Tissue Eng Regen Med 2022; 16:961-976. [PMID: 36112528 PMCID: PMC9826348 DOI: 10.1002/term.3349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/05/2022] [Accepted: 09/01/2022] [Indexed: 01/12/2023]
Abstract
Activation of the canonical Wingless-related integration site (Wnt) pathway has been shown to increase bone formation and therefore has therapeutic potential for use in orthopedic conditions. However, attempts at developing an effective strategy to achieve Wnt activation has been met with several challenges. The inherent hydrophobicity of Wnt ligands makes isolating and purifying the protein difficult. To circumvent these challenges, many have sought to target extracellular inhibitors of the Wnt pathway, such as Wnt signaling pathway inhibitors Sclerostin and Dickkopf-1, or to use small molecules, ions and proteins to increase target Wnt genes. Here, we review systemic and localized bioactive approaches to enhance bone formation or improve bone repair through antibody-based therapeutics, synthetic Wnt surrogates and scaffold doping to target canonical Wnt. We conclude with a brief review of emerging technologies, such as mRNA therapy and Clustered Regularly Interspaced Short Palindromic Repeats technology, which serve as promising approaches for future clinical translation.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA
| | - GianLuca Fontana
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Elizabeth Miclau
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA
| | - Mallory Rongstad
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - William Murphy
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA,Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Johnny Huard
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Nicole Ehrhart
- School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Chelsea Bahney
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA,Orthopaedic Trauma InstituteUniversity of California, San Francisco (UCSF)San FranciscoCaliforniaUSA
| |
Collapse
|
15
|
Targeted therapy in Coronavirus disease 2019 (COVID-19): Implication from cell and gene therapy to immunotherapy and vaccine. Int Immunopharmacol 2022; 111:109161. [PMID: 35998506 PMCID: PMC9385778 DOI: 10.1016/j.intimp.2022.109161] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) is a highly pathogenic and transmissible virus. Infection caused by SARS-CoV-2 known as Coronavirus disease 2019 (COVID-19) can be severe, especially among high risk populations affected of underlying medical conditions. COVID-19 is characterized by the severe acute respiratory syndrome, a hyper inflammatory syndrome, vascular injury, microangiopathy and thrombosis. Antiviral drugs and immune modulating methods has been evaluated. So far, a particular therapeutic option has not been approved for COVID-19 and a variety of treatments have been studied for COVID-19 including, current treatment such as oxygen therapy, corticosteroids, antiviral agents until targeted therapy and vaccines which are diverse in each patient and have various outcomes. According to the findings of different in vitro and in vivo studies, some novel approach such as gene editing, cell based therapy, and immunotherapy may have significant potential in the treatment of COVID-19. Based on these findings, this paper aims to review the different strategies of treatment against COVID-19 and provide a summary from traditional and newer methods in curing COVID-19.
Collapse
|
16
|
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: 5.0] [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
|
17
|
Wu J, Vunjak-Novakovic G. Bioengineering human cartilage-bone tissues for modeling of osteoarthritis. Stem Cells Dev 2022; 31:399-405. [PMID: 35088600 PMCID: PMC9398485 DOI: 10.1089/scd.2021.0317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease worldwide, yet we continue to lack an understanding of disease etiology and pathology, and effective treatment options. Essential to tissue homeostasis, disease pathogenesis, and therapeutic responses are the stratified organization of cartilage and the crosstalk at the osteochondral junction. Animal models may capture some of these features, but to establish clinically consistent therapeutics, there remains a need for high-fidelity models of OA that meet all the above requirements in a human, patient-specific manner. In vitro bioengineered cartilage-bone tissue models could be developed to recapitulate physiological interactions with human cells and disease initiating factors. Here we highlight human induced pluripotent stem cells (hiPSCs) as the advantageous cell source for these models and review approaches for chondrogenic fate specification from hiPSCs. To achieve native-like stratified cartilage organization with cartilage-bone interactions, spatiotemporal cues mimicking development can be delivered to engineered tissues by patterning of the cells, scaffold, and the environment. Once healthy and native-like cartilage-bone tissues are established, an OA-like state can be induced via cytokine challenge or injurious loading. Bioengineered cartilage-bone tissues fall short of recapitulating the full complexity of native tissues, but have demonstrated utility in elucidating some mechanisms of OA progression and enabled screening of candidate therapeutics in patient-specific models. With rapid progress in stem cells, tissue engineering, imaging, and high throughput -omics research in recent years, we propose that advanced human tissue models will soon offer valuable contributions to our understanding and treatment of OA.
Collapse
Affiliation(s)
- Josephine Wu
- Columbia University, 5798, Biomedical Engineering, New York, New York, United States;
| | - Gordana Vunjak-Novakovic
- Columbia University, 5798, Biomedical Engineering, 622 west 168th St, VC12-234, New York, New York, United States, 10032;
| |
Collapse
|
18
|
Lee MH, Shin JI, Yang JW, Lee KH, Cha DH, Hong JB, Park Y, Choi E, Tizaoui K, Koyanagi A, Jacob L, Park S, Kim JH, Smith L. Genome Editing Using CRISPR-Cas9 and Autoimmune Diseases: A Comprehensive Review. Int J Mol Sci 2022; 23:1337. [PMID: 35163260 PMCID: PMC8835887 DOI: 10.3390/ijms23031337] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Autoimmune diseases are disorders that destruct or disrupt the body's own tissues by its own immune system. Several studies have revealed that polymorphisms of multiple genes are involved in autoimmune diseases. Meanwhile, gene therapy has become a promising approach in autoimmune diseases, and clustered regularly interspaced palindromic repeats and CRISPR-associated protein 9 (CRISPR-Cas9) has become one of the most prominent methods. It has been shown that CRISPR-Cas9 can be applied to knock out proprotein convertase subtilisin/kexin type 9 (PCSK9) or block PCSK9, resulting in lowering low-density lipoprotein cholesterol. In other studies, it can be used to treat rare diseases such as ornithine transcarbamylase (OTC) deficiency and hereditary tyrosinemia. However, few studies on the treatment of autoimmune disease using CRISPR-Cas9 have been reported so far. In this review, we highlight the current and potential use of CRISPR-Cas9 in the management of autoimmune diseases. We summarize the potential target genes for immunomodulation using CRISPR-Cas9 in autoimmune diseases including rheumatoid arthritis (RA), inflammatory bowel diseases (IBD), systemic lupus erythematosus (SLE), multiple sclerosis (MS), type 1 diabetes mellitus (DM), psoriasis, and type 1 coeliac disease. This article will give a new perspective on understanding the use of CRISPR-Cas9 in autoimmune diseases not only through animal models but also in human models. Emerging approaches to investigate the potential target genes for CRISPR-Cas9 treatment may be promising for the tailored immunomodulation of some autoimmune diseases in the near future.
Collapse
Affiliation(s)
- Min Ho Lee
- Yonsei University College of Medicine, Seoul 03722, Korea; (M.H.L.); (D.H.C.); (J.B.H.); (Y.P.); (E.C.); (S.P.)
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (J.I.S.); (K.H.L.)
| | - Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea;
| | - Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (J.I.S.); (K.H.L.)
| | - Do Hyeon Cha
- Yonsei University College of Medicine, Seoul 03722, Korea; (M.H.L.); (D.H.C.); (J.B.H.); (Y.P.); (E.C.); (S.P.)
- Korea Advanced Institute for Science and Technology, Graduate School of Medical Science and Engineering, Daejeon 34141, Korea
| | - Jun Beom Hong
- Yonsei University College of Medicine, Seoul 03722, Korea; (M.H.L.); (D.H.C.); (J.B.H.); (Y.P.); (E.C.); (S.P.)
| | - Yeoeun Park
- Yonsei University College of Medicine, Seoul 03722, Korea; (M.H.L.); (D.H.C.); (J.B.H.); (Y.P.); (E.C.); (S.P.)
| | - Eugene Choi
- Yonsei University College of Medicine, Seoul 03722, Korea; (M.H.L.); (D.H.C.); (J.B.H.); (Y.P.); (E.C.); (S.P.)
| | - Kalthoum Tizaoui
- Laboratory Microorganismes and Active Biomolecules, Sciences Faculty of Tunis, University Tunis El Manar, Tunis 1068, Tunisia;
| | - Ai Koyanagi
- Parc Sanitari Sant Joan de Deu/CIBERSAM, Universitat de Barcelona, Fundacio Sant Joan de Deu, Sant Boi de Llobregat, 08830 Barcelona, Spain; (A.K.); (L.J.)
- ICREA, Pg. LluisCompanys 23, 08010 Barcelona, Spain
| | - Louis Jacob
- Parc Sanitari Sant Joan de Deu/CIBERSAM, Universitat de Barcelona, Fundacio Sant Joan de Deu, Sant Boi de Llobregat, 08830 Barcelona, Spain; (A.K.); (L.J.)
- Faculty of Medicine, University of Versailles Saint-Quentin-en-Yvelines, 78180 Montigny-le-Bretonneux, France
| | - Seoyeon Park
- Yonsei University College of Medicine, Seoul 03722, Korea; (M.H.L.); (D.H.C.); (J.B.H.); (Y.P.); (E.C.); (S.P.)
| | - Ji Hong Kim
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Lee Smith
- Centre for Health, Performance, and Wellbeing, Anglia Ruskin University, Cambridge CB1 1PT, UK;
| |
Collapse
|
19
|
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: 9.0] [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
|
20
|
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: 21] [Impact Index Per Article: 7.0] [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
|
21
|
Cahill SV, Kwon HK, Back J, Lee I, Lee S, Alder KD, Hao Z, Yu KE, Dussik CM, Kyriakides TR, Lee FY. Locally delivered adjuvant biofilm-penetrating antibiotics rescue impaired endochondral fracture healing caused by MRSA infection. J Orthop Res 2021; 39:402-414. [PMID: 33336805 DOI: 10.1002/jor.24965] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 02/04/2023]
Abstract
Infection is a devastating complication following an open fracture. We investigated whether local rifampin-loaded hydrogel can combat infection and improve healing in a murine model of methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis. A transverse fracture was made at the tibia midshaft of C57BL/6J mice aged 10-12 weeks and stabilized with an intramedullary pin. A total of 1 × 106 colony-forming units (CFU) of MRSA was inoculated. A collagen-based hydrogel containing low-dose (60 μg) and high-dose (300 μg) rifampin was applied before closure. Postoperative treatment response was assessed through bacterial CFU counts from tissue and hardware, tibial radiographs and microcomputed tomography (μCT), immunohistochemistry, and histological analyses. All untreated MRSA-infected fractures progressed to nonunion by 28 days with profuse MRSA colonization. Infected fractures demonstrated decreased soft callus formation on safranin O stain compared to controls. Areas of dense interleukin-1β stain were associated with poor callus formation. High-dose rifampin hydrogels reduced the average MRSA load in tissue (p < 0.0001) and implants (p = 0.041). Low-dose rifampin hydrogels reduced tissue bacterial load by 50% (p = 0.021). Among sterile models, 88% achieved union compared to 0% of those infected. Mean radiographic union scale in tibia scores improved from 6 to 8.7 with high-dose rifampin hydrogel (p = 0.024) and to 10 with combination local/systemic rifampin therapy (p < 0.0001). μCT demonstrated reactive bone formation in MRSA infection. Histology demonstrated restored fracture healing with bacterial elimination. Rifampin-loaded hydrogels suppressed osteomyelitis, prevented implant colonization, and improved healing. Systemic rifampin was more effective at eliminating infection and improving fracture healing. Further investigation into rifampin-loaded hydrogels is required to correlate these findings with clinical efficacy.
Collapse
Affiliation(s)
- Sean V Cahill
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jungho Back
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| | - Inkyu Lee
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Saelim Lee
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
- College of Medicine, Dankook University, Yongin, Gyeonggi-do, Republic of Korea
| | - Kareme D Alder
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| | - Zichen Hao
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Kristin E Yu
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christopher M Dussik
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
22
|
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: 1.0] [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
|
23
|
De Pieri A, Byerley AM, Musumeci CR, Salemizadehparizi F, Vanderhorst MA, Wuertz‐Kozak K. Electrospinning and 3D bioprinting for intervertebral disc tissue engineering. JOR Spine 2020; 3:e1117. [PMID: 33392454 PMCID: PMC7770193 DOI: 10.1002/jsp2.1117] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a major cause of low back pain and represents a massive socioeconomic burden. Current conservative and surgical treatments fail to restore native tissue architecture and functionality. Tissue engineering strategies, especially those based on 3D bioprinting and electrospinning, have emerged as possible alternatives by producing cell-seeded scaffolds that replicate the structure of the IVD extracellular matrix. In this review, we provide an overview of recent advancements and limitations of 3D bioprinting and electrospinning for the treatment of IVD degeneration, focusing on future areas of research that may contribute to their clinical translation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | - Ann M. Byerley
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | - Catherine R. Musumeci
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | | | - Maya A. Vanderhorst
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
| | - Karin Wuertz‐Kozak
- Department of Biomedical EngineeringRochester Institute of Technology (RIT)RochesterNew YorkUSA
- Schön Clinic Munich Harlaching, Spine CenterAcademic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (AU)MunichGermany
| |
Collapse
|
24
|
Caobi A, Dutta RK, Garbinski LD, Esteban-Lopez M, Ceyhan Y, Andre M, Manevski M, Ojha CR, Lapierre J, Tiwari S, Parira T, El-Hage N. The Impact of CRISPR-Cas9 on Age-related Disorders: From Pathology to Therapy. Aging Dis 2020; 11:895-915. [PMID: 32765953 PMCID: PMC7390517 DOI: 10.14336/ad.2019.0927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
With advances in medical technology, the number of people over the age of 60 is on the rise, and thus, increasing the prevalence of age-related pathologies within the aging population. Neurodegenerative disorders, cancers, metabolic and inflammatory diseases are some of the most prevalent age-related pathologies affecting the growing population. It is imperative that a new treatment to combat these pathologies be developed. Although, still in its infancy, the CRISPR-Cas9 system has become a potent gene-editing tool capable of correcting gene-mediated age-related pathology, and therefore ameliorating or eliminating disease symptoms. Deleting target genes using the CRISPR-Cas9 system or correcting for gene mutations may ameliorate many different neurodegenerative disorders detected in the aging population. Cancer cells targeted by the CRISPR-Cas9 system may result in an increased sensitivity to chemotherapeutics, lower proliferation, and higher cancer cell death. Finally, reducing gene targeting inflammatory molecules production through microRNA knockout holds promise as a therapeutic strategy for both arthritis and inflammation. Here we present a review based on how the expanding world of genome editing can be applied to disorders and diseases affecting the aging population.
Collapse
Affiliation(s)
- Allen Caobi
- 1Departments of Immunology and Nano-medicine
| | | | - Luis D Garbinski
- 3Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Huynh NP, Gloss CC, Lorentz J, Tang R, Brunger JM, McAlinden A, Zhang B, Guilak F. Long non-coding RNA GRASLND enhances chondrogenesis via suppression of the interferon type II signaling pathway. eLife 2020; 9:49558. [PMID: 32202492 PMCID: PMC7202894 DOI: 10.7554/elife.49558] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 03/21/2020] [Indexed: 12/14/2022] Open
Abstract
The roles of long noncoding RNAs (lncRNAs) in musculoskeletal development, disease, and regeneration remain poorly understood. Here, we identified the novel lncRNA GRASLND (originally named RNF144A-AS1) as a regulator of mesenchymal stem cell (MSC) chondrogenesis. GRASLND, a primate-specific lncRNA, is upregulated during MSC chondrogenesis and appears to act directly downstream of SOX9, but not TGF-β3. We showed that the silencing of GRASLND resulted in lower accumulation of cartilage-like extracellular matrix in a pellet assay, while GRASLND overexpression – either via transgene ectopic expression or by endogenous activation via CRISPR-dCas9-VP64 – significantly enhanced cartilage matrix production. GRASLND acts to inhibit IFN-γ by binding to EIF2AK2, and we further demonstrated that GRASLND exhibits a protective effect in engineered cartilage against interferon type II. Our results indicate an important role of GRASLND in regulating stem cell chondrogenesis, as well as its therapeutic potential in the treatment of cartilage-related diseases, such as osteoarthritis.
Collapse
Affiliation(s)
- Nguyen Pt Huynh
- Department of Orthopaedic Surgery, Washington University, St Louis, United States.,Shriners Hospitals for Children, St. Louis, United States.,Department of Cell Biology, Duke University, Durham, United States.,Center of Regenerative Medicine, Washington University, St Louis, United States
| | - Catherine C Gloss
- Department of Orthopaedic Surgery, Washington University, St Louis, United States.,Shriners Hospitals for Children, St. Louis, United States.,Center of Regenerative Medicine, Washington University, St Louis, United States
| | - Jeremiah Lorentz
- Department of Orthopaedic Surgery, Washington University, St Louis, United States.,Shriners Hospitals for Children, St. Louis, United States.,Center of Regenerative Medicine, Washington University, St Louis, United States
| | - Ruhang Tang
- Department of Orthopaedic Surgery, Washington University, St Louis, United States.,Shriners Hospitals for Children, St. Louis, United States.,Center of Regenerative Medicine, Washington University, St Louis, United States
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, United States
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University, St Louis, United States.,Shriners Hospitals for Children, St. Louis, United States.,Center of Regenerative Medicine, Washington University, St Louis, United States
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University, St Louis, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St Louis, United States.,Shriners Hospitals for Children, St. Louis, United States.,Center of Regenerative Medicine, Washington University, St Louis, United States
| |
Collapse
|
26
|
Abstract
Connective tissues within the synovial joints are characterized by their dense extracellular matrix and sparse cellularity. With injury or disease, however, tissues commonly experience an influx of cells owing to proliferation and migration of endogenous mesenchymal cell populations, as well as invasion of the tissue by other cell types, including immune cells. Although this process is critical for successful wound healing, aberrant immune-mediated cell infiltration can lead to pathological inflammation of the joint. Importantly, cells of mesenchymal or haematopoietic origin use distinct modes of migration and thus might respond differently to similar biological cues and microenvironments. Furthermore, cell migration in the physiological microenvironment of musculoskeletal tissues differs considerably from migration in vitro. This Review addresses the complexities of cell migration in fibrous connective tissues from three separate but interdependent perspectives: physiology (including the cellular and extracellular factors affecting 3D cell migration), pathophysiology (cell migration in the context of synovial joint autoimmune disease and injury) and tissue engineering (cell migration in engineered biomaterials). Improved understanding of the fundamental mechanisms governing interstitial cell migration might lead to interventions that stop invasion processes that culminate in deleterious outcomes and/or that expedite migration to direct endogenous cell-mediated repair and regeneration of joint tissues.
Collapse
|
27
|
Pferdehirt L, Ross AK, Brunger JM, Guilak F. A Synthetic Gene Circuit for Self-Regulating Delivery of Biologic Drugs in Engineered Tissues. Tissue Eng Part A 2019; 25:809-820. [PMID: 30968743 DOI: 10.1089/ten.tea.2019.0027] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
IMPACT STATEMENT We engineered a synthetic transcription system based on nuclear factor kappa-light-chain-enhancer of activated B cells signaling that can attenuate the effects of the inflammatory cytokine interleukin (IL)-1α in a self-regulating manner. This system responds in a time- and dose-dependent manner to rapidly produce therapeutic levels of IL-1 receptor antagonist (IL-1Ra). The use of lentiviral gene therapy allows this system to be utilized through different transduction methods and in different cell types for a variety of applications. Broadly, this approach may be applicable in developing autoregulated biologic systems for tissue engineering and drug delivery in a range of disease applications.
Collapse
Affiliation(s)
- Lara Pferdehirt
- 1 Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,3 Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri.,4 Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Alison K Ross
- 1 Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,3 Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri.,4 Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Jonathan M Brunger
- 5 Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - Farshid Guilak
- 1 Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,3 Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri.,4 Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| |
Collapse
|
28
|
陈 果, 程 度, 陈 滨. [Development of CRISPR technology and its application in bone and cartilage tissue engineering]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1515-1520. [PMID: 31907146 PMCID: PMC6942994 DOI: 10.12122/j.issn.1673-4254.2019.12.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 12/09/2022]
Abstract
The CRISPR/Cas9 system, consisting of Cas9 nuclease and single guide RNA (sgRNA), is an emerging gene editing technology that can perform gene reprogramming operations such as deletion, insertion, and point mutation on DNA sequences targeted by sgRNA. In addition, CRISPR/dCas9 (a mutant that loses Cas9 nuclease activity) still retains the ability of sgRNA to target DNA. The fusion of dCas9 protein with transcriptional activator (CRISPRa) can activate the expression of the target gene, and fusion transcriptional repressors (CRISPRi) can also be used to suppress target gene expression. Efficient delivery of the CRISPR/Cas9 system is one of the main problems limiting its wide clinical application. Viral vectors are widely used to efficiently deliver CRISPR/Cas9 elements, but non-viral vector research is more attractive in terms of safety, simplicity, and flexibility. In this review, we summarize the principles and research advances of CRISPR technology, including CRISPR/ Cas9 delivery vectors, delivery methods, and obstacles to the delivery, and review the progress of CRISPR-based research in bone and cartilage tissue engineering. Finally, the challenges and future applications of CRISPR technology in bone and cartilage tissue engineering are discussed.
Collapse
Affiliation(s)
- 果 陈
- 南方医科大学南方医院创伤骨科,广东 广州 510515Department of Traumatology and Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 度 程
- 中山大学材料科学与工程学院,广东 广州 510275School of Material Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - 滨 陈
- 南方医科大学南方医院创伤骨科,广东 广州 510515Department of Traumatology and Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
29
|
Khorraminejad-Shirazi M, Dorvash M, Estedlal A, Hoveidaei AH, Mazloomrezaei M, Mosaddeghi P. Aging: A cell source limiting factor in tissue engineering. World J Stem Cells 2019; 11:787-802. [PMID: 31692986 PMCID: PMC6828594 DOI: 10.4252/wjsc.v11.i10.787] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/03/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering has yet to reach its ideal goal, i.e. creating profitable off-the-shelf tissues and organs, designing scaffolds and three-dimensional tissue architectures that can maintain the blood supply, proper biomaterial selection, and identifying the most efficient cell source for use in cell therapy and tissue engineering. These are still the major challenges in this field. Regarding the identification of the most appropriate cell source, aging as a factor that affects both somatic and stem cells and limits their function and applications is a preventable and, at least to some extents, a reversible phenomenon. Here, we reviewed different stem cell types, namely embryonic stem cells, adult stem cells, induced pluripotent stem cells, and genetically modified stem cells, as well as their sources, i.e. autologous, allogeneic, and xenogeneic sources. Afterward, we approached aging by discussing the functional decline of aged stem cells and different intrinsic and extrinsic factors that are involved in stem cell aging including replicative senescence and Hayflick limit, autophagy, epigenetic changes, miRNAs, mTOR and AMPK pathways, and the role of mitochondria in stem cell senescence. Finally, various interventions for rejuvenation and geroprotection of stem cells are discussed. These interventions can be applied in cell therapy and tissue engineering methods to conquer aging as a limiting factor, both in original cell source and in the in vitro proliferated cells.
Collapse
Affiliation(s)
- Mohammadhossein Khorraminejad-Shirazi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mohammadreza Dorvash
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Alireza Estedlal
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Amir Human Hoveidaei
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mohsen Mazloomrezaei
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Pouria Mosaddeghi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Cell and Molecular Medicine Student Research Group, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| |
Collapse
|
30
|
RNA-based therapy for osteogenesis. Int J Pharm 2019; 569:118594. [DOI: 10.1016/j.ijpharm.2019.118594] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 02/06/2023]
|
31
|
CRISPR technologies for stem cell engineering and regenerative medicine. Biotechnol Adv 2019; 37:107447. [PMID: 31513841 DOI: 10.1016/j.biotechadv.2019.107447] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
CRISPR/Cas9 system exploits the concerted action of Cas9 nuclease and programmable single guide RNA (sgRNA), and has been widely used for genome editing. The Cas9 nuclease activity can be abolished by mutation to yield the catalytically deactivated Cas9 (dCas9). Coupling with the customizable sgRNA for targeting, dCas9 can be fused with transcription repressors to inhibit specific gene expression (CRISPR interference, CRISPRi) or fused with transcription activators to activate the expression of gene of interest (CRISPR activation, CRISPRa). Here we introduce the principles and recent advances of these CRISPR technologies, their delivery vectors and review their applications in stem cell engineering and regenerative medicine. In particular, we focus on in vitro stem cell fate manipulation and in vivo applications such as prevention of retinal and muscular degeneration, neural regeneration, bone regeneration, cartilage tissue engineering, as well as treatment of diseases in blood, skin and liver. Finally, the challenges to translate CRISPR to regenerative medicine and future perspectives are discussed and proposed.
Collapse
|
32
|
Farhang N, Ginley-Hidinger M, Berrett KC, Gertz J, Lawrence B, Bowles RD. Lentiviral CRISPR Epigenome Editing of Inflammatory Receptors as a Gene Therapy Strategy for Disc Degeneration. Hum Gene Ther 2019; 30:1161-1175. [PMID: 31140325 PMCID: PMC6761595 DOI: 10.1089/hum.2019.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
Degenerative disc disease (DDD) is a primary contributor to low-back pain, a leading cause of disability. Progression of DDD is aided by inflammatory cytokines in the intervertebral disc (IVD), particularly TNF-α and IL-1β, but current treatments fail to effectively target this mechanism. The objective of this study was to explore the feasibility of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) epigenome editing-based therapy for DDD, by modulation of TNFR1/IL1R1 signaling in pathological human IVD cells. Human IVD cells from the nucleus pulposus of patients receiving surgery for back pain were obtained and the regulation of TNFR1/IL1R1 signaling by a lentiviral CRISPR epigenome editing system was tested. These cells were tested for successful lentiviral transduction/expression of deactivated Cas9 fused to Krüppel Associated Box system and regulation of TNFR1/IL1R1 expression. TNFR1/IL1R1 signaling disruption was investigated through measurement of NF-κB activity, apoptosis, and anabolic/catabolic changes in gene expression postinflammatory challenge. CRISPR epigenome editing systems were effectively introduced into pathological human IVD cells and significantly downregulated TNFR1 and IL1R1. This downregulation significantly attenuated deleterious TNFR1 signaling but not IL1R1 signaling. This is attributed to less robust IL1R1 expression downregulation, and IL-1β-driven reversal of IL1R1 expression downregulation in a portion of patient IVD cells. In addition, RNAseq data indicated novel transcription factor targets, IRF1 and TFAP2C, as being primary regulators of inflammatory signaling in IVD cells. These results demonstrate the feasibility of CRISPR epigenome editing of inflammatory receptors in pathological IVD cells, but highlight a limitation in epigenome targeting of IL1R1. This method has potential application as a novel gene therapy for DDD, to attenuate the deleterious effect of inflammatory cytokines present in the degenerative IVD.
Collapse
MESH Headings
- Apoptosis
- Biomarkers
- Cells, Cultured
- Clustered Regularly Interspaced Short Palindromic Repeats
- Epigenesis, Genetic
- Gene Editing
- Gene Expression Regulation
- Gene Order
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Vectors/genetics
- Humans
- Intervertebral Disc Degeneration/genetics
- Intervertebral Disc Degeneration/therapy
- Lentivirus/genetics
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Interleukin-1 Type I/genetics
- Receptors, Interleukin-1 Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Transduction, Genetic
Collapse
Affiliation(s)
- Niloofar Farhang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
| | | | | | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
| | - Brandon Lawrence
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| | - Robby D. Bowles
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
33
|
Choi YR, Collins KH, Lee JW, Kang HJ, Guilak F. Genome Engineering for Osteoarthritis: From Designer Cells to Disease-Modifying Drugs. Tissue Eng Regen Med 2019; 16:335-343. [PMID: 31413938 PMCID: PMC6675820 DOI: 10.1007/s13770-018-0172-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/27/2018] [Accepted: 12/01/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a highly prevalent degenerative joint disease involving joint cartilage and its surrounding tissues. OA is the leading cause of pain and disability worldwide. At present, there are no disease-modifying OA drugs, and the primary therapies include exercise and nonsteroidal anti-inflammatory drugs until total joint replacement at the end-stage of the disease. METHODS In this review, we summarized the current state of knowledge in genetic and epigenetic associations and risk factors for OA and their potential diagnostic and therapeutic applications. RESULTS Genome-wide association studies and analysis of epigenetic modifications (such as miRNA expression, DNA methylation and histone modifications) conducted across various populations support the notion that there is a genetic basis for certain subsets of OA pathogenesis. CONCLUSION With recent advances in the development of genome editing technologies such as the CRISPR-Cas9 system, these genetic and epigenetic alternations in OA can be used as platforms from which potential biomarkers for the diagnosis, prognosis, drug response, and development of potential personalized therapeutic targets for OA can be approached. Furthermore, genome editing has allowed the development of "designer" cells, whereby the receptors, gene regulatory networks, or transgenes can be modified as a basis for new cell-based therapies.
Collapse
Affiliation(s)
- Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO 63130 USA
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis, MO 63110 USA
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Republic of Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO 63130 USA
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis, MO 63110 USA
| | - Jin-Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Republic of Korea
| | - Ho-Jung Kang
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Republic of Korea
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO 63130 USA
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis, MO 63110 USA
- Center of Regenerative Medicine, Campus Box 8233, McKinley Research Bldg, Room 3121, St. Louis, MO 63110 USA
| |
Collapse
|
34
|
Ewart D, Peterson EJ, Steer CJ. A new era of genetic engineering for autoimmune and inflammatory diseases. Semin Arthritis Rheum 2019; 49:e1-e7. [DOI: 10.1016/j.semarthrit.2019.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023]
|
35
|
Wong RS, Chen YY, Smolke CD. Regulation of T cell proliferation with drug-responsive microRNA switches. Nucleic Acids Res 2019; 46:1541-1552. [PMID: 29244152 PMCID: PMC5815133 DOI: 10.1093/nar/gkx1228] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/30/2017] [Indexed: 11/21/2022] Open
Abstract
As molecular and cellular therapies advance in the clinic, the role of genetic regulation is becoming increasingly important for controlling therapeutic potency and safety. The emerging field of mammalian synthetic biology provides promising tools for the construction of regulatory platforms that can intervene with endogenous pathways and control cell behavior. Recent work has highlighted the development of synthetic biological systems that integrate sensing of molecular signals to regulated therapeutic function in various disease settings. However, the toxicity and limited dosing of currently available molecular inducers have largely inhibited translation to clinical settings. In this work, we developed synthetic microRNA-based genetic systems that are controlled by the pharmaceutical drug leucovorin, which is readily available and safe for prolonged administration in clinical settings. We designed microRNA switches to target endogenous cytokine receptor subunits (IL-2Rβ and γc) that mediate various signaling pathways in T cells. We demonstrate the function of these control systems by effectively regulating T cell proliferation with the drug input. Each control system produced unique functional responses, and combinatorial targeting of multiple receptor subunits exhibited greater repression of cell growth. This work highlights the potential use of drug-responsive genetic control systems to improve the management and safety of cellular therapeutics.
Collapse
Affiliation(s)
- Remus S Wong
- Department of Bioengineering, 443 Via Ortega, MC 4245, Stanford University, Stanford, CA 94305, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, 420 Westwood Plaza, Boelter Hall 5531, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Christina D Smolke
- Department of Bioengineering, 443 Via Ortega, MC 4245, Stanford University, Stanford, CA 94305, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
36
|
Nguyen AH, Marsh P, Schmiess-Heine L, Burke PJ, Lee A, Lee J, Cao H. Cardiac tissue engineering: state-of-the-art methods and outlook. J Biol Eng 2019; 13:57. [PMID: 31297148 PMCID: PMC6599291 DOI: 10.1186/s13036-019-0185-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to assess the state-of-the-art fabrication methods, advances in genome editing, and the use of machine learning to shape the prospective growth in cardiac tissue engineering. Those interdisciplinary emerging innovations would move forward basic research in this field and their clinical applications. The long-entrenched challenges in this field could be addressed by novel 3-dimensional (3D) scaffold substrates for cardiomyocyte (CM) growth and maturation. Stem cell-based therapy through genome editing techniques can repair gene mutation, control better maturation of CMs or even reveal its molecular clock. Finally, machine learning and precision control for improvements of the construct fabrication process and optimization in tissue-specific clonal selections with an outlook of cardiac tissue engineering are also presented.
Collapse
Affiliation(s)
- Anh H. Nguyen
- Electrical and Computer Engineering Department, University of Alberta, Edmonton, Alberta Canada
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Paul Marsh
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Lauren Schmiess-Heine
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Peter J. Burke
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Chemical Engineering and Materials Science Department, University of California Irvine, Irvine, CA USA
| | - Abraham Lee
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Mechanical and Aerospace Engineering Department, University of California Irvine, Irvine, CA USA
| | - Juhyun Lee
- Bioengineering Department, University of Texas at Arlington, Arlington, TX USA
| | - Hung Cao
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Henry Samueli School of Engineering, University of California, Irvine, USA
| |
Collapse
|
37
|
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: 20] [Impact Index Per Article: 4.0] [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
|
38
|
Loibl M, Wuertz‐Kozak K, Vadala G, Lang S, Fairbank J, Urban JP. Controversies in regenerative medicine: Should intervertebral disc degeneration be treated with mesenchymal stem cells? JOR Spine 2019; 2:e1043. [PMID: 31463457 PMCID: PMC6711491 DOI: 10.1002/jsp2.1043] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/31/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022] Open
Abstract
Low back pain (LBP) can significantly reduce the quality of life of patients, and has a considerable economic and social impact worldwide. It is commonly associated with disc degeneration, even though many people with degenerate discs are asymptomatic. Degenerate disc disease (DDD), is thus a common term for intervertebral disc (IVD) degeneration associated with LBP. Degeneration is thought to lead to LBP because of nerve ingrowth into the degenerate disc, inflammation, or because degradation of extracellular matrix (ECM) alters spinal biomechanics inappropriately. Thus, while the objectives of some interventions for LBP are to control pain intensity, other interventions aim to deal with the consequences of disc degeneration through stabilizing the disc surgically, by inserting artificial discs or by repairing the disc biologically and preventing progressive IVD degeneration. Despite tremendous research efforts, treatment of LBP through the use of regenerative interventions aiming to repair the IVD is still controversial. The use of mesenchymal stem cells for IVD regeneration in a patient-based case will be discussed by an ensemble of clinicians and researchers.
Collapse
Affiliation(s)
- Markus Loibl
- Department of Spine SurgerySchulthess KlinikZürichSwitzerland
- Department of Trauma SurgeryRegensburg University Medical CenterRegensburgGermany
| | - Karin Wuertz‐Kozak
- Institute for Biomechanics, Department of Health Sciences and TechnologyETH Zürich, ZürichSwitzerland
- Spine Center, Schön Klinik MünchenMunichGermany
- Academic Teaching Hospital and Spine Research InstituteParacelsus Private Medical UniversitySalzburgAustria
- Department of Health ScienceUniversity of PotsdamPotsdamGermany
| | - Gianluca Vadala
- Department of Orthopaedic and Trauma SurgeryCampus Bio‐Medico University of RomeRomeItaly
| | - Siegmund Lang
- Department of Trauma SurgeryRegensburg University Medical CenterRegensburgGermany
| | - Jeremy Fairbank
- Nuffield Department of OrthopaedicsRheumatology and Musculoskeletal Sciences (NDORMS), University of OxfordOxfordUK
| | - Jill P. Urban
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
39
|
Buduru SD, Gulei D, Zimta AA, Tigu AB, Cenariu D, Berindan-Neagoe I. The Potential of Different Origin Stem Cells in Modulating Oral Bone Regeneration Processes. Cells 2019; 8:cells8010029. [PMID: 30625993 PMCID: PMC6356555 DOI: 10.3390/cells8010029] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 12/22/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering has gained much momentum since the implementation of stem cell isolation and manipulation for regenerative purposes. Despite significant technical improvements, researchers still have to decide which strategy (which type of stem cell) is the most suitable for their specific purpose. Therefore, this short review discusses the advantages and disadvantages of the three main categories of stem cells: embryonic stem cells, mesenchymal stem cells and induced pluripotent stem cells in the context of bone regeneration for dentistry-associated conditions. Importantly, when deciding upon the right strategy, the selection needs to be made in concordance with the morbidity and the life-threatening level of the condition in discussion. Therefore, even when a specific type of stem cell holds several advantages over others, their availability, invasiveness of the collection method and ethical standards become deciding parameters.
Collapse
Affiliation(s)
- Smaranda Dana Buduru
- Prosthetics and Dental Materials, Faculty of Dental Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, 32 Clinicilor Street, 400006 Cluj-Napoca, Romania.
- Stomestet Stomatology Clinic, Calea Manastur 68A Street, 400658 Cluj-Napoca, Romania; .
| | - Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Adrian Bogdan Tigu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Diana Cenariu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Republicii 34-36 Street, 400015 Cluj-Napoca, Romania.
| |
Collapse
|
40
|
Lolli A, Colella F, De Bari C, van Osch GJVM. Targeting anti-chondrogenic factors for the stimulation of chondrogenesis: A new paradigm in cartilage repair. J Orthop Res 2019; 37:12-22. [PMID: 30175861 DOI: 10.1002/jor.24136] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/09/2018] [Indexed: 02/04/2023]
Abstract
Trauma and age-related cartilage disorders represent a major global cause of morbidity, resulting in chronic pain and disability in patients. A lack of effective therapies, together with a rapidly aging population, creates an impressive clinical and economic burden on healthcare systems. In this scenario, experimental therapies based on transplantation or in situ stimulation of skeletal Mesenchymal Stem/progenitor Cells (MSCs) have raised great interest for cartilage repair. Nevertheless, the challenge of guiding MSC differentiation and preventing cartilage hypertrophy and calcification still needs to be overcome. While research has mostly focused on the stimulation of cartilage anabolism using growth factors, several issues remain unresolved prompting the field to search for novel solutions. Recently, inhibition of anti-chondrogenic regulators has emerged as an intriguing opportunity. Anti-chondrogenic regulators include extracellular proteins as well as intracellular transcription factors and microRNAs that act as potent inhibitors of pro-chondrogenic signals. Suppression of these inhibitors can enhance MSC chondrogenesis and production of cartilage matrix. We here review the current knowledge concerning different types of anti-chondrogenic regulators. We aim to highlight novel therapeutic targets for cartilage repair and discuss suitable tools for suppressing their anti-chondrogenic functions. Further effort is needed to unveil the therapeutic perspectives of this approach and pave the way for effective treatment of cartilage injuries in patients. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands
| | - Fabio Colella
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands.,Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
41
|
Allen MJ. What's New in Musculoskeletal Basic Science. J Bone Joint Surg Am 2018; 100:2082-2086. [PMID: 30516632 DOI: 10.2106/jbjs.18.01055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Matthew J Allen
- Department of Veterinary Medicine, Surgical Discovery Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
42
|
Biomaterial-guided delivery of gene vectors for targeted articular cartilage repair. Nat Rev Rheumatol 2018; 15:18-29. [DOI: 10.1038/s41584-018-0125-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Graceffa V, Vinatier C, Guicheux J, Stoddart M, Alini M, Zeugolis DI. Chasing Chimeras - The elusive stable chondrogenic phenotype. Biomaterials 2018; 192:199-225. [PMID: 30453216 DOI: 10.1016/j.biomaterials.2018.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
Abstract
The choice of the best-suited cell population for the regeneration of damaged or diseased cartilage depends on the effectiveness of culture conditions (e.g. media supplements, three-dimensional scaffolds, mechanical stimulation, oxygen tension, co-culture systems) to induce stable chondrogenic phenotype. Herein, advances and shortfalls in in vitro, preclinical and clinical setting of various in vitro microenvironment modulators on maintaining chondrocyte phenotype or directing stem cells towards chondrogenic lineage are critically discussed. Chondrocytes possess low isolation efficiency, limited proliferative potential and rapid phenotypic drift in culture. Mesenchymal stem cells are relatively readily available, possess high proliferation potential, exhibit great chondrogenic differentiation capacity, but they tend to acquire a hypertrophic phenotype when exposed to chondrogenic stimuli. Embryonic and induced pluripotent stem cells, despite their promising in vitro and preclinical data, are still under-investigated. Although a stable chondrogenic phenotype remains elusive, recent advances in in vitro microenvironment modulators are likely to develop clinically- and commercially-relevant therapies in the years to come.
Collapse
Affiliation(s)
- Valeria Graceffa
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Claire Vinatier
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Jerome Guicheux
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Martin Stoddart
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
44
|
Graceffa V, Vinatier C, Guicheux J, Evans CH, Stoddart M, Alini M, Zeugolis DI. State of art and limitations in genetic engineering to induce stable chondrogenic phenotype. Biotechnol Adv 2018; 36:1855-1869. [DOI: 10.1016/j.biotechadv.2018.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
|
45
|
Ede D, Davidoff N, Blitch A, Farhang N, Bowles RD. Microfluidic Flow Cell Array for Controlled Cell Deposition in Engineered Musculoskeletal Tissues. Tissue Eng Part C Methods 2018; 24:546-556. [DOI: 10.1089/ten.tec.2018.0184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- David Ede
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | | | - Alejandro Blitch
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Niloofar Farhang
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Robby D. Bowles
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
- Department of Orthopedics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
46
|
Ewart DT, Peterson EJ, Steer CJ. Gene editing for inflammatory disorders. Ann Rheum Dis 2018; 78:6-15. [PMID: 30077989 DOI: 10.1136/annrheumdis-2018-213454] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 12/24/2022]
Abstract
Technology for precise and efficient genetic editing is constantly evolving and is now capable of human clinical applications. Autoimmune and inflammatory diseases are chronic, disabling, sometimes life-threatening, conditions that feature heritable components. Both primary genetic lesions and the inflammatory pathobiology underlying these diseases represent fertile soil for new therapies based on the capabilities of gene editing. The ability to orchestrate precise targeted modifications to the genome will likely enable cell-based therapies for inflammatory diseases such as monogenic autoinflammatory disease, acquired autoimmune disease and for regenerative medicine in the setting of an inflammatory environment. Here, we discuss recent advances in genome editing and their evolving applications in immunoinflammatory diseases. Strengths and limitations of older genetic modification tools are compared with CRISPR/Cas9, base editing, RNA editing, targeted activators and repressors of transcription and targeted epigenetic modifiers. Commonly employed delivery vehicles to target cells or tissues of interest with genetic modification machinery, including viral, non-viral and cellular vectors, are described. Finally, applications in animal and human models of inflammatory diseases are discussed. Use of chimeric autoantigen receptor T cells, correction of monogenic diseases with genetically edited haematopoietic stem and progenitor cells, engineering of induced pluripotent stem cells and ex vivo expansion and modification of regulatory T cells for a range of chronic inflammatory diseases are reviewed.
Collapse
Affiliation(s)
- David T Ewart
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Erik J Peterson
- Division of Rheumatic and Autoimmune Diseases, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Clifford J Steer
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
47
|
Krupkova O, Cambria E, Besse L, Besse A, Bowles R, Wuertz‐Kozak K. The potential of CRISPR/Cas9 genome editing for the study and treatment of intervertebral disc pathologies. JOR Spine 2018; 1:e1003. [PMID: 31463435 PMCID: PMC6686831 DOI: 10.1002/jsp2.1003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/09/2018] [Accepted: 02/12/2018] [Indexed: 12/11/2022] Open
Abstract
The CRISPR/Cas9 system has emerged as a powerful tool for mammalian genome engineering. In basic and translational intervertebral disc (IVD) research, this technique has remarkable potential to answer fundamental questions on pathway interactions, to simulate IVD pathologies, and to promote drug development. Furthermore, the precisely targeted CRISPR/Cas9 gene therapy holds promise for the effective and targeted treatment of degenerative disc disease and low back pain. In this perspective, we provide an overview of recent CRISPR/Cas9 advances stemming from/with transferability to IVD research, outline possible treatment approaches for degenerative disc disease, and discuss current limitations that may hinder clinical translation.
Collapse
Affiliation(s)
- Olga Krupkova
- Department of Health Sciences and TechnologyInstitute for BiomechanicsETH ZurichSwitzerland
| | - Elena Cambria
- Department of Health Sciences and TechnologyInstitute for BiomechanicsETH ZurichSwitzerland
| | - Lenka Besse
- Department of Oncology and HematologyCantonal Hospital St GallenSt GallenSwitzerland
| | - Andrej Besse
- Department of Oncology and HematologyCantonal Hospital St GallenSt GallenSwitzerland
| | - Robert Bowles
- Department of BioengineeringUniversity of UtahSalt Lake CityUtah
- Department of OrthopaedicsUniversity of UtahSalt Lake CityUtah
| | - Karin Wuertz‐Kozak
- Department of Health Sciences and TechnologyInstitute for BiomechanicsETH ZurichSwitzerland
- Spine CenterSchön Klinik München HarlachingMunichGermany
- Academic Teaching Hospital and Spine Research InstituteParacelsus Private Medical University SalzburgSalzburgAustria
- Department of Health SciencesUniversity of PotsdamPotsdamGermany
| |
Collapse
|
48
|
Giau VV, Lee H, Shim KH, Bagyinszky E, An SSA. Genome-editing applications of CRISPR-Cas9 to promote in vitro studies of Alzheimer's disease. Clin Interv Aging 2018; 13:221-233. [PMID: 29445268 PMCID: PMC5808714 DOI: 10.2147/cia.s155145] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Genetic variations play an important role in the clinical presentation and progression of Alzheimer’s disease (AD), especially early-onset Alzheimer’s disease. Hundreds of mutations have been reported with the majority resulting from alterations in β-amyloid precursor protein (APP), presenilin 1 (PSEN1), or presenilin 2 (PSEN2) genes. The roles of these mutations in the pathogenesis of AD have been classically confirmed or refuted through functional studies, where the mutations are cloned, inserted into cell lines, and monitored for changes in various properties including cell survival, amyloid production, or Aβ42/40 ratio. However, these verification studies tend to be expensive, time consuming, and inconsistent. Recently, the clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR–Cas9) system was developed, which improves sequence-specific gene editing in cell lines, organs, and animals. CRISPR–Cas9 is a promising tool for the generation of models of human genetic diseases and could facilitate the establishment of new animal AD models and the observation of dynamic bioprocesses in AD. Here, we recapitulated the history of CRISPR technology, recent progress, and, especially, its potential applications in AD-related genetic, animal modeling, and functional studies.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Hyon Lee
- Department of Neurology, Gachon University Gil Medical Center, Incheon, South Korea
| | - Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Eva Bagyinszky
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| |
Collapse
|
49
|
Adkar SS, Brunger JM, Willard VP, Wu CL, Gersbach CA, Guilak F. Genome Engineering for Personalized Arthritis Therapeutics. Trends Mol Med 2017; 23:917-931. [PMID: 28887050 PMCID: PMC5657581 DOI: 10.1016/j.molmed.2017.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023]
Abstract
Arthritis represents a family of complex joint pathologies responsible for the majority of musculoskeletal conditions. Nearly all diseases within this family, including osteoarthritis, rheumatoid arthritis, and juvenile idiopathic arthritis, are chronic conditions with few or no disease-modifying therapeutics available. Advances in genome engineering technology, most recently with CRISPR-Cas9, have revolutionized our ability to interrogate and validate genetic and epigenetic elements associated with chronic diseases such as arthritis. These technologies, together with cell reprogramming methods, including the use of induced pluripotent stem cells, provide a platform for human disease modeling. We summarize new evidence from genome-wide association studies and genomics that substantiates a genetic basis for arthritis pathogenesis. We also review the potential contributions of genome engineering in the development of new arthritis therapeutics.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Chia-Lung Wu
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Cytex Therapeutics, Inc., Durham, NC 27705, USA.
| |
Collapse
|
50
|
Farhang N, Brunger JM, Stover JD, Thakore PI, Lawrence B, Guilak F, Gersbach CA, Setton LA, Bowles RD. * CRISPR-Based Epigenome Editing of Cytokine Receptors for the Promotion of Cell Survival and Tissue Deposition in Inflammatory Environments. Tissue Eng Part A 2017; 23:738-749. [PMID: 28095751 PMCID: PMC5568019 DOI: 10.1089/ten.tea.2016.0441] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/11/2017] [Indexed: 01/08/2023] Open
Abstract
Musculoskeletal diseases have been associated with inflammatory cytokine action, particularly action by TNF-α and IL-1β. These inflammatory cytokines promote apoptosis and senescence of cells in diseased tissue and extracellular matrix breakdown. Stem cell-based therapies are being considered for the treatment of musculoskeletal diseases, but the presence of these inflammatory cytokines will have similar deleterious action on therapeutic cells delivered to these environments. Methods that prevent inflammatory-induced apoptosis and proinflammatory signaling, in cell and pathway-specific manners are needed. In this study we demonstrate the use of clustered regularly interspaced short palindromic repeats (CRISPR)-based epigenome editing to alter cell response to inflammatory environments by repressing inflammatory cytokine cell receptors, specifically TNFR1 and IL1R1. We targeted CRISPR/Cas9-based repressors to TNFR1 and IL1R1 gene regulatory elements in human adipose-derived stem cells (hADSCs) and investigated the functional outcomes of repression of these genes. Efficient signaling regulation was demonstrated in engineered hADSCs, as activity of the downstream transcription factor NF-κB was significantly reduced or maintained at baseline levels in the presence of TNF-α or IL-1β. Pellet culture of undifferentiated hADSCs demonstrated improved survival in engineered hADSCs treated with TNF-α or IL-1β, while having little effect on their immunomodulatory properties. Furthermore, engineered hADSCs demonstrated improved chondrogenic differentiation capacity in the presence of TNF-α or IL-1β, as shown by superior production of glycosaminglycans in this inflammatory environment. Overall this work demonstrates a novel method for modulating cell response to inflammatory signaling that has applications in engineering cells delivered to inflammatory environments, and as a direct gene therapy to protect endogenous cells exposed to chronic inflammation, as observed in a broad spectrum of degenerative musculoskeletal pathology.
Collapse
Affiliation(s)
- Niloofar Farhang
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Joshua D. Stover
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | | | - Brandon Lawrence
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| | - Farshid Guilak
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri
- Department of Orthopaedic Surgery, Washington University in St. Louis and Shriners Hospitals for Children–St. Louis, Saint Louis, Missouri
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Lori A. Setton
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri
- Department of Orthopaedic Surgery, Washington University in St. Louis and Shriners Hospitals for Children–St. Louis, Saint Louis, Missouri
| | - Robby D. Bowles
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| |
Collapse
|