1
|
Lee AJ, Gangi LR, Hu YJ, Dinescu AT, Guo XE, Bozynski CC, Kuroki K, Stoker AM, Marra KG, Ateshian GA, Cook JL, Hung CT. Evaluation of Dexamethasone-Eluting Cell-Seeded Constructs in a Preclinical Canine Model of Cartilage Repair. Tissue Eng Part A 2025; 31:208-218. [PMID: 39607494 PMCID: PMC11971544 DOI: 10.1089/ten.tea.2024.0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
In this 12-month long, preclinical large animal study using a canine model, we report that engineered osteochondral grafts (comprised of allogeneic chondrocyte-seeded hydrogels with the capacity for sustained release of the corticosteroid dexamethasone [DEX], cultured to functional mechanical properties, and incorporated over porous titanium bases), can successfully repair damaged cartilage. DEX release from within engineered cartilage was hypothesized to improve initial cartilage repair by modulating the local inflammatory environment, which was also associated with suppressed degenerative changes exhibited by menisci and synovium. We note that not all histological and clinical outcomes at an intermediary time point of three months paralleled 12-month outcomes, which emphasizes the importance of in vivo studies in valid preclinical models that incorporate clinically relevant follow-up durations. Together, our study demonstrates that engineered cartilage fabricated under the conditions reported herein can repair full-thickness cartilage defects and promote synovial joint health and function.
Collapse
Affiliation(s)
- Andy J. Lee
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Lianna R. Gangi
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Yizhong Jenny Hu
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Andreea T. Dinescu
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - X. Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Chantelle C. Bozynski
- Thompson Laboratory for Regenerative Orthopaedics, Missouri Orthopaedic Institute, Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, USA
| | - Keiichi Kuroki
- Thompson Laboratory for Regenerative Orthopaedics, Missouri Orthopaedic Institute, Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, USA
| | - Aaron M. Stoker
- Thompson Laboratory for Regenerative Orthopaedics, Missouri Orthopaedic Institute, Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, USA
| | - Kacey G. Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- Department of Mechanical Engineering, Columbia University, New York, New York, USA
| | - James L. Cook
- Thompson Laboratory for Regenerative Orthopaedics, Missouri Orthopaedic Institute, Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, USA
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| |
Collapse
|
2
|
Gangi LR, Pagon AD, Pellicore MJ, Kroupa KR, Murphy LA, Ateshian GA, Hung CT. Synovium friction properties are influenced by proteoglycan content. J Biomech 2024; 174:112272. [PMID: 39146899 PMCID: PMC11825662 DOI: 10.1016/j.jbiomech.2024.112272] [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: 05/10/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
The synovium plays a crucial role in diarthrodial joint health, and its study has garnered appreciation as synovitis has been linked to osteoarthritis symptoms and progression. Quantitative synovium structure-function data, however, remain sparse. In the present study, we hypothesized that tissue glycosaminoglycan (GAG) content contributes to the low friction properties of the synovium. Bovine and human synovium tribological properties were evaluated using a custom friction testing device in two different cases: (1) proteoglycan depletion to isolate the influence of tissue GAGs in the synovium friction response and (2) interleukin-1 (IL) treatment to observe inflammation-induced structural and functional changes. Following proteoglycan depletion, synovium friction coefficients increased while GAG content decreased. Conversely, synovium explants treated with the proinflammatory cytokine IL exhibited elevated GAG concentrations and decreased friction coefficients. For the first time, a relationship between synovium friction coefficient and GAG concentration is demonstrated. The study of synovium tribology is necessary to fully understand the mechanical environment of the healthy and diseased joint.
Collapse
Affiliation(s)
- Lianna R Gangi
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Athena D Pagon
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Matthew J Pellicore
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Kimberly R Kroupa
- Department of Mechanical Engineering, Columbia University, New York, NY, United States
| | - Lance A Murphy
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, New York, NY, United States; Department of Mechanical Engineering, Columbia University, New York, NY, United States
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, New York, NY, United States; Department of Orthopedic Surgery, Columbia University, New York, NY, United States.
| |
Collapse
|
3
|
Marrero - Berrios I, Salter SE, Hirday R, Rabolli CP, Tan A, Hung CT, Schloss RS, Yarmush ML. In vitro inflammatory multi-cellular model of osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100432. [PMID: 38288345 PMCID: PMC10823137 DOI: 10.1016/j.ocarto.2023.100432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Objective Osteoarthritis (OA) is a chronic joint disease, with limited treatment options, characterized by inflammation and matrix degradation, and resulting in severe pain or disability. Progressive inflammatory interaction among key cell types, including chondrocytes and macrophages, leads to a cascade of intra- and inter-cellular events which culminate in OA induction. In order to investigate these interactions, we developed a multi-cellular in vitro OA model, to characterize OA progression, and identify and evaluate potential OA therapeutics in response to mediators representing graded levels of inflammatory severity. Methods We compared macrophages, chondrocytes and their co-culture responses to "low" Interleukin-1 (IL-1) or "high" IL-1/tumor necrosis factor (IL-1/TNF) levels of inflammation. We also investigated response changes following the administration of dexamethasone (DEX) or mesenchymal stromal cell (MSC) treatment via a combination of gene expression and secretory changes, reflecting not only inflammation, but also chondrocyte function. Results Inflamed chondrocytes presented an osteoarthritic-like phenotype characterized by high gene expression of pro-inflammatory cytokines and chemokines, up-regulation of ECM degrading proteases, and down-regulation of chondrogenic genes. Our results indicate that while MSC treatment attenuates macrophage inflammation directly, it does not reduce chondrocyte inflammatory responses, unless macrophages are present as well. DEX however, can directly attenuate chondrocyte inflammation. Conclusions Our results highlight the importance of considering multi-cellular interactions when studying complex systems such as the articular joint. In addition, our approach, using a panel of both inflammatory and chondrocyte functional genes, provides a more comprehensive approach to investigate disease biomarkers, and responses to treatment.
Collapse
Affiliation(s)
| | - S. Elina Salter
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Rishabh Hirday
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Charles P. Rabolli
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Andrea Tan
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Rene S. Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Martin L. Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
4
|
Luzzi AJ, Ferrer X, Fang F, Golman M, Song L, Marshall BP, Lee AJ, Kim JJ, Hung CT, Thomopoulos S. Hedgehog Activation for Enhanced Rotator Cuff Tendon-to-Bone Healing. Am J Sports Med 2023; 51:3825-3834. [PMID: 37897335 PMCID: PMC10821775 DOI: 10.1177/03635465231203210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/30/2023]
Abstract
BACKGROUND Rotator cuff repair is a common orthopaedic procedure, yet the rate of failure to heal after surgery is high. Repair site rupture is due to poor tendon-to-bone healing and lack of regeneration of the native fibrocartilaginous enthesis. During development, the enthesis is formed and mineralized by a pool of progenitors activated by hedgehog signaling. Furthermore, hedgehog signaling drives regenerative enthesis healing in young animals, in contrast to older animals, in which enthesis injuries heal via fibrovascular scar and without participation of hedgehog signaling. HYPOTHESIS Hedgehog activation improves tendon-to-bone healing in an animal model of rotator cuff repair. STUDY DESIGN Controlled laboratory study. METHODS A total of 78 adult Sprague-Dawley rats were used. Supraspinatus tendon injury and repair were completed bilaterally, with microsphere-encapsulated hedgehog agonist administered to right shoulders and control microspheres administered to left shoulders. Animals were sacrificed after 3, 14, 28, or 56 days. Gene expression and histological, biomechanical, and bone morphometric analyses were conducted. RESULTS At 3 days, hedgehog signaling pathway genes Gli1 (1.70; P = .029) and Smo (2.06; P = .0173), as well as Runx2 (1.69; P = .0386), a transcription factor of osteogenesis, were upregulated in treated relative to control repairs. At 14 days, transcription factors of tenogenesis, Scx (4.00; P = .041), and chondrogenesis, Sox9 (2.95; P = .010), and mineralized fibrocartilage genes Col2 (3.18; P = .031) and Colx (1.85; P = .006), were upregulated in treated relative to control repairs. Treatment promoted fibrocartilage formation at the healing interface by 28 days, with improvements in tendon-bone maturity, organization, and continuity. Treatment led to improved biomechanical properties. The material property strength (2.43 vs 1.89 N/m2; P = .046) and the structural property work to failure (29.01 vs 18.09 mJ; P = .030) were increased in treated relative to control repairs at 28 days and 56 days, respectively. Treatment had a marginal effect on bone morphometry underlying the repair. Trabecular thickness (0.08 vs 0.07 mm; P = .035) was increased at 28 days. CONCLUSION Hedgehog agonist treatment activated hedgehog signaling at the tendon-to-bone repair site and prompted increased mineralized fibrocartilage production. This extracellular matrix production and mineralization resulted in improved biomechanical properties, demonstrating the therapeutic potential of hedgehog agonism for improving tendon-to-bone healing after rotator cuff repair. CLINICAL RELEVANCE This study demonstrates the therapeutic potential of hedgehog agonist treatment for improving tendon-to-bone healing after rotator cuff injury and repair.
Collapse
Affiliation(s)
- Andrew J. Luzzi
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
| | - Xavier Ferrer
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
| | - Fei Fang
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
| | - Mikhail Golman
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Lee Song
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
| | - Brittany P. Marshall
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Andy J. Lee
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Jieon J. Kim
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
| | - Clark T. Hung
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Stavros Thomopoulos
- Department of Orthopaedic Surgery, Columbia University, New York, New York, USA
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| |
Collapse
|
5
|
Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels: An emerging therapeutic strategy for cartilage regeneration. Adv Colloid Interface Sci 2023; 321:103030. [PMID: 37907031 DOI: 10.1016/j.cis.2023.103030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Martyn Snow
- Department of Arthroscopy, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham B31 2AP, United Kingdom; The Robert Jones and Agnes Hunt Hospital, Oswestry, Shropshire SY10 7AG, United Kingdom
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom; Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Pooya Davoodi
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom.
| |
Collapse
|
6
|
Lee AJ, Gangi LR, Zandkarimi F, Stockwell BR, Hung CT. Red blood cell exposure increases chondrocyte susceptibility to oxidative stress following hemarthrosis. Osteoarthritis Cartilage 2023; 31:1365-1376. [PMID: 37364817 PMCID: PMC10529126 DOI: 10.1016/j.joca.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/11/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023]
Abstract
OBJECTIVE The detrimental effects of blood exposure on articular tissues are well characterized, but the individual contributions of specific whole blood components are yet to be fully elucidated. Better understanding of mechanisms that drive cell and tissue damage in hemophilic arthropathy will inform novel therapeutic strategies. The studies here aimed to identify the specific contributions of intact and lysed red blood cells (RBCs) on cartilage and the therapeutic potential of Ferrostatin-1 in the context of lipid changes, oxidative stress, and ferroptosis. METHODS Changes to biochemical and mechanical properties following intact RBC treatment were assessed in human chondrocyte-based tissue-engineered cartilage constructs and validated against human cartilage explants. Chondrocyte monolayers were assayed for changes to intracellular lipid profiles and the presence of oxidative and ferroptotic mechanisms. RESULTS Markers of tissue breakdown were observed in cartilage constructs without parallel losses in DNA (control: 786.3 (102.2) ng/mg; RBCINT: 751 (126.4) ng/mg; P = 0.6279), implicating nonlethal chondrocyte responses to intact RBCs. Dose-dependent loss of viability in response to intact and lysed RBCs was observed in chondrocyte monolayers, with greater toxicity observed with lysates. Intact RBCs induced changes to chondrocyte lipid profiles, upregulating highly oxidizable fatty acids (e.g., FA 18:2) and matrix disrupting ceramides. RBC lysates induced cell death via oxidative mechanisms that resemble ferroptosis. CONCLUSIONS Intact RBCs induce intracellular phenotypic changes to chondrocytes that increase vulnerability to tissue damage while lysed RBCs have a more direct influence on chondrocyte death by mechanisms that are representative of ferroptosis.
Collapse
Affiliation(s)
- Andy J Lee
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, USA.
| | - Lianna R Gangi
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, USA.
| | - Fereshteh Zandkarimi
- Department of Chemistry, Columbia University, 216 Havemeyer Hall, 3000 Broadway, Mail Code 3183, New York, NY, USA.
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, 216 Havemeyer Hall, 3000 Broadway, Mail Code 3183, New York, NY, USA; Department of Biological Sciences, Columbia University, 1208 NWC Building, 550 West 120th St. M.C. 4846, New York, NY, USA.
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, USA; Department of Orthopaedic Surgery, Columbia University, New York, NY, USA.
| |
Collapse
|
7
|
Marshall BP, Ferrer XE, Kunes JA, Innis AC, Luzzi AJ, Forrester LA, Burt KG, Lee AJ, Song L, Hung CT, Levine WN, Kovacevic D, Thomopoulos S. The subacromial bursa is a key regulator of the rotator cuff and a new therapeutic target for improving repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.547347. [PMID: 37425730 PMCID: PMC10327214 DOI: 10.1101/2023.07.01.547347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Rotator cuff injuries result in over 500,000 surgeries performed annually, an alarmingly high number of which fail. These procedures typically involve repair of the injured tendon and removal of the subacromial bursa. However, recent identification of a resident population of mesenchymal stem cells and inflammatory responsiveness of the bursa to tendinopathy indicate an unexplored biological role of the bursa in the context of rotator cuff disease. Therefore, we aimed to understand the clinical relevance of bursa-tendon crosstalk, characterize the biologic role of the bursa within the shoulder, and test the therapeutic potential for targeting the bursa. Proteomic profiling of patient bursa and tendon samples demonstrated that the bursa is activated by tendon injury. Using a rat to model rotator cuff injury and repair, tenotomy-activated bursa protected the intact tendon adjacent to the injured tendon and maintained the morphology of the underlying bone. The bursa also promoted an early inflammatory response in the injured tendon, initiating key players in wound healing. In vivo results were supported by targeted organ culture studies of the bursa. To examine the potential to therapeutically target the bursa, dexamethasone was delivered to the bursa, prompting a shift in cellular signaling towards resolution of inflammation in the healing tendon. In conclusion, contrary to current clinical practice, the bursa should be retained to the greatest extent possible and provides a new therapeutically target for improving tendon healing outcomes. One Sentence Summary The subacromial bursa is activated by rotator cuff injury and regulates the paracrine environment of the shoulder to maintain the properties of the underlying tendon and bone.
Collapse
|
8
|
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: 2.5] [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
|
9
|
Madamsetty VS, Mohammadinejad R, Uzieliene I, Nabavi N, Dehshahri A, García-Couce J, Tavakol S, Moghassemi S, Dadashzadeh A, Makvandi P, Pardakhty A, Aghaei Afshar A, Seyfoddin A. Dexamethasone: Insights into Pharmacological Aspects, Therapeutic Mechanisms, and Delivery Systems. ACS Biomater Sci Eng 2022; 8:1763-1790. [PMID: 35439408 PMCID: PMC9045676 DOI: 10.1021/acsbiomaterials.2c00026] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dexamethasone (DEX) has been widely used to treat a variety of diseases, including autoimmune diseases, allergies, ocular disorders, cancer, and, more recently, COVID-19. However, DEX usage is often restricted in the clinic due to its poor water solubility. When administered through a systemic route, it can elicit severe side effects, such as hypertension, peptic ulcers, hyperglycemia, and hydro-electrolytic disorders. There is currently much interest in developing efficient DEX-loaded nanoformulations that ameliorate adverse disease effects inhibiting advancements in scientific research. Various nanoparticles have been developed to selectively deliver drugs without destroying healthy cells or organs in recent years. In the present review, we have summarized some of the most attractive applications of DEX-loaded delivery systems, including liposomes, polymers, hydrogels, nanofibers, silica, calcium phosphate, and hydroxyapatite. This review provides our readers with a broad spectrum of nanomedicine approaches to deliver DEX safely.
Collapse
Affiliation(s)
- Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida 32224, United States
| | - Reza Mohammadinejad
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman 7618866749, Iran
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406 Vilnius, Lithuania
| | - Noushin Nabavi
- Department of Urologic Sciences, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Jomarien García-Couce
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
- Department of Polymeric Biomaterials, Biomaterials Center (BIOMAT), University of Havana, Havana 10600, Cuba
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1417755469, Iran
| | - Saeid Moghassemi
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7618866748, Iran
| | - Abbas Aghaei Afshar
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman 7618866749, Iran
| | - Ali Seyfoddin
- Drug Delivery Research Group, Auckland University of Technology (AUT), School of Science, Auckland 1010, New Zealand
| |
Collapse
|
10
|
Wang X, Li X, Duffy P, McMahon S, Wang X, Lyu J, Xu Q, A S, Chen NN, Bi V, Dürig T, Wang W. Resveratrol‐Loaded Poly(
d
,
l
‐Lactide‐
Co
‐Glycolide) Microspheres Integrated in a Hyaluronic Acid Injectable Hydrogel for Cartilage Regeneration. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Xi Wang
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
- Ashland Specialties Ireland Ltd. National Science Park Building V, Dublin Road, Petitswood, Mullingar Co. Westmeath N91 F6PD Ireland
| | - Xiaolin Li
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
- Ashland Specialties Ireland Ltd. National Science Park Building V, Dublin Road, Petitswood, Mullingar Co. Westmeath N91 F6PD Ireland
| | - Patrick Duffy
- Ashland Specialties Ireland Ltd. National Science Park Building V, Dublin Road, Petitswood, Mullingar Co. Westmeath N91 F6PD Ireland
| | - Sean McMahon
- Ashland Specialties Ireland Ltd. National Science Park Building V, Dublin Road, Petitswood, Mullingar Co. Westmeath N91 F6PD Ireland
| | - Xianqing Wang
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
| | - Jing Lyu
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
| | - Qian Xu
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
| | - Sigen A
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
| | - Ningyi N. Chen
- Pharmaceutical R&D Ashland Specialty Ingredients G.P. 500 Hercules Road, 8136A/260 Wilmington DE 19808 USA
| | - Vivian Bi
- Pharmaceutical R&D Ashland Specialty Ingredients G.P. 500 Hercules Road, 8136A/260 Wilmington DE 19808 USA
| | - Thomas Dürig
- Pharmaceutical R&D Ashland Specialty Ingredients G.P. 500 Hercules Road, 8136A/260 Wilmington DE 19808 USA
| | - Wenxin Wang
- Charles Institute of Dermatology School of Medicine University College Dublin Dublin 4 Ireland
| |
Collapse
|
11
|
Gupta N, Kamath S M, Rao SK, D J, Patil S, Gupta N, Arunachalam KD. Kaempferol loaded albumin nanoparticles and dexamethasone encapsulation into electrospun polycaprolactone fibrous mat – Concurrent release for cartilage regeneration. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Lee AJ, Mahoney CM, Cai CC, Ichinose R, Stefani RM, Marra KG, Ateshian GA, Shah RP, Vunjak-Novakovic G, Hung CT. Sustained Delivery of SB-431542, a Type I Transforming Growth Factor Beta-1 Receptor Inhibitor, to Prevent Arthrofibrosis. Tissue Eng Part A 2021; 27:1411-1421. [PMID: 33752445 DOI: 10.1089/ten.tea.2021.0029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fibrosis of the knee is a common disorder resulting from an aberrant wound healing response and is characterized by extracellular matrix deposition, joint contraction, and scar tissue formation. The principal regulator of the fibrotic cascade is transforming growth factor beta-1 (TGF-β1), a factor that induces rapid proliferation and differentiation of resident fibroblasts. In this study, we demonstrate successful inhibition of TGF-β1-driven myofibroblastic differentiation in human fibroblast-like synoviocytes using a small molecule TGF-β1 receptor inhibitor, SB-431542. We also demonstrate successful encapsulation of SB-431542 in poly(D,L-lactide-co-glycolide) (PLGA) as a potential prophylactic treatment for arthrofibrosis and characterize drug release and bioactivity in a three-dimensional collagen gel contraction assay. We assessed the effects of TGF-β1 and SB-431542 on cell proliferation and viability in monolayer cultures. Opposing dose-dependent trends were observed in cell proliferation, which increased in TGF-β1-treated cultures and decreased in SB-431542-treated cultures relative to control (p < 0.05). SB-431542 was not cytotoxic at the concentrations studied (0-50 μM) and inhibited TGF-β1-induced collagen gel contraction in a dose-dependent manner. Specifically, TGF-β1-treated gels contracted to 18% ± 1% of their initial surface area, while gels treated with TGF-β1 and ≥10 μM SB-431542 showed no evidence of contraction (p < 0.0001). Upon removal of the compound, all gels contracted to control levels after 44 h in culture, necessitating sustained delivery for prolonged inhibition. To this end, SB-431542 was encapsulated in PLGA microspheres (SBMS) that had an average diameter of 87.5 ± 24 μm and a loading capacity of 4.3 μg SB-431542 per milligram of SBMS. Functional assessment of SBMS revealed sustained inhibition of TGF-β1-induced gel contraction as well as hallmark features of myofibroblastic differentiation, including α-smooth muscle actin expression and connective tissue growth factor production. These results suggest that SB-431542 may be used to counter TGF-β1-driven events in the fibrotic cascade in the knee cartilage.
Collapse
Affiliation(s)
- Andy J Lee
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Christopher M Mahoney
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles C Cai
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Rika Ichinose
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Robert M Stefani
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York, USA.,Department of Mechanical Engineering, Columbia University, New York, New York, USA
| | - Roshan P Shah
- Department of Orthopaedic Surgery, and Columbia University, New York, New York, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, New York, USA.,Department of Medicine, Columbia University, New York, New York, USA
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, New York, New York, USA.,Department of Orthopaedic Surgery, and Columbia University, New York, New York, USA
| |
Collapse
|
13
|
Rahmani Del Bakhshayesh A, Babaie S, Tayefi Nasrabadi H, Asadi N, Akbarzadeh A, Abedelahi A. An overview of various treatment strategies, especially tissue engineering for damaged articular cartilage. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:1089-1104. [DOI: 10.1080/21691401.2020.1809439] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Azizeh Rahmani Del Bakhshayesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soraya Babaie
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Stefani RM, Lee AJ, Tan AR, Halder SS, Hu Y, Guo XE, Stoker AM, Ateshian GA, Marra KG, Cook JL, Hung CT. Sustained low-dose dexamethasone delivery via a PLGA microsphere-embedded agarose implant for enhanced osteochondral repair. Acta Biomater 2020; 102:326-340. [PMID: 31805408 PMCID: PMC6956850 DOI: 10.1016/j.actbio.2019.11.052] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022]
Abstract
Articular cartilage defects are a common source of joint pain and dysfunction. We hypothesized that sustained low-dose dexamethasone (DEX) delivery via an acellular osteochondral implant would have a dual pro-anabolic and anti-catabolic effect, both supporting the functional integrity of adjacent graft and host tissue while also attenuating inflammation caused by iatrogenic injury. An acellular agarose hydrogel carrier with embedded DEX-loaded poly(lactic-co-glycolic) acid (PLGA) microspheres (DLMS) was developed to provide sustained release for at least 99 days. The DLMS implant was first evaluated in an in vitro pro-inflammatory model of cartilage degradation. The implant was chondroprotective, as indicated by maintenance of Young's modulus (EY) (p = 0.92) and GAG content (p = 1.0) in the presence of interleukin-1β insult. In a subsequent preliminary in vivo experiment, an osteochondral autograft transfer was performed using a pre-clinical canine model. DLMS implants were press-fit into the autograft donor site and compared to intra-articular DEX injection (INJ) or no DEX (CTL). Functional scores for DLMS animals returned to baseline (p = 0.39), whereas CTL and INJ remained significantly worse at 6 months (p < 0.05). DLMS knees were significantly more likely to have improved OARSI scores for proteoglycan, chondrocyte, and collagen pathology (p < 0.05). However, no significant improvements in synovial fluid cytokine content were observed. In conclusion, utilizing a targeted DLMS implant, we observed in vitro chondroprotection in the presence of IL-1-induced degradation and improved in vivo functional outcomes. These improved outcomes were correlated with superior histological scores but not necessarily a dampened inflammatory response, suggesting a primarily pro-anabolic effect. STATEMENT OF SIGNIFICANCE: Articular cartilage defects are a common source of joint pain and dysfunction. Effective treatment of these injuries may prevent the progression of osteoarthritis and reduce the need for total joint replacement. Dexamethasone, a potent glucocorticoid with concomitant anti-catabolic and pro-anabolic effects on cartilage, may serve as an adjuvant for a variety of repair strategies. Utilizing a dexamethasone-loaded osteochondral implant with controlled release characteristics, we demonstrated in vitro chondroprotection in the presence of IL-1-induced degradation and improved in vivo functional outcomes following osteochondral repair. These improved outcomes were correlated with superior histological cartilage scores and minimal-to-no comorbidity, which is a risk with high dose dexamethasone injections. Using this model of cartilage restoration, we have for the first time shown the application of targeted, low-dose dexamethasone for improved healing in a preclinical model of focal defect repair.
Collapse
Affiliation(s)
- Robert M Stefani
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Andy J Lee
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Andrea R Tan
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Saiti S Halder
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Aaron M Stoker
- Missouri Orthopaedic Institute, University of Missouri, 1100 Virginia Avenue, Columbia 65212, MO, United States
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States; Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York 10027, NY, United States
| | - Kacey G Marra
- University of Pittsburgh, Biomedical Science Tower, 200 Lothrop Street, Pittsburgh 15213, PA, United States
| | - James L Cook
- Missouri Orthopaedic Institute, University of Missouri, 1100 Virginia Avenue, Columbia 65212, MO, United States
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States.
| |
Collapse
|
15
|
Connizzo BK, Grodzinsky AJ. Lose-Dose Administration of Dexamethasone Is Beneficial in Preventing Secondary Tendon Damage in a Stress-Deprived Joint Injury Explant Model. J Orthop Res 2020; 38:139-149. [PMID: 31441099 PMCID: PMC7268908 DOI: 10.1002/jor.24451] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/13/2019] [Indexed: 02/04/2023]
Abstract
Secondary joint damage is the process by which a single injury can lead to detrimental changes in adjacent tissue structures, typically through the spread of inflammatory responses. We recently developed an in vitro model of secondary joint damage using a murine rotator cuff explant system, in which injuries to muscle and bone cause massive cell death in otherwise uninjured tendon. The purpose of the present study was to test the ability cytokine-targeted and broad-spectrum therapeutics to prevent cell death and tissue degeneration associated with secondary joint damage. We treated injured bone-tendon-muscle explants with either interleukin-1 receptor antagonist, etanercept, or dexamethasone (DEX) for up to 7 days in culture. Only the low-dose DEX treatment was able to prevent cell death and tissue degeneration. We then identified a critical window between 24 and 72 h following injury for maximal benefit of DEX treatment through timed administration experiments. Finally, we performed two tendon-only explant studies to identify mechanistic effects on tendon health. Interestingly, DEX did not prevent cell death and degeneration in a model of cytokine-induced damage, suggesting other targets of DEX activity. Future studies will aim to identify factors in joint inflammation that may be targeted by DEX treatment, as well as to investigate novel delivery strategies. Statement of clinical significance: Overall, this work demonstrates beneficial effects of DEX administration on preventing tenocyte death and extracellular matrix degeneration in an explant model of secondary joint damage, supporting the clinical use of low-dose glucocorticoids for short-term treatment of joint inflammation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:139-149, 2020.
Collapse
Affiliation(s)
- Brianne K. Connizzo
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, United States,Correspondence: Brianne K. Connizzo,
70 Massachusetts Avenue, NE47-377, Cambridge, MA 02139, T: 617-253-2469,
| | - Alan J. Grodzinsky
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, United States,Department of Electrical Engineering and Computer Science,
Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, United States
| |
Collapse
|
16
|
Du HL, Zhai AD, Yu H. Synergistic effect of halofuginone and dexamethasone on LPS‑induced acute lung injury in type II alveolar epithelial cells and a rat model. Mol Med Rep 2019; 21:927-935. [PMID: 31974595 DOI: 10.3892/mmr.2019.10865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 11/09/2018] [Indexed: 11/09/2022] Open
Abstract
Acute lung injury (ALI) is characterized by neutrophilic infiltration, uncontrolled oxidative stress and inflammatory processes. Despite various therapeutic regimes having been performed, there remains no effective pharmacotherapy available to treat ALI. Halofuginone (HF), a ketone isolated from Dichroa febrifuga, exhibits significant anti‑inflammatory and antifibrotic effects. Dexamethasone (DEX), a synthetic glucocorticoid, has been routinely used as an adjuvant therapy in treating inflammatory diseases, including ALI. The present study aimed to investigate the effects of the combination of HF and DEX in the treatment of ALI. The present results suggested that the simultaneous administration of HF and DEX markedly decreased the level of pro‑inflammatory cytokines and increased the level of anti‑inflammatory cytokines, as assessed by western blot analysis. In addition, HF and DEX effectively decreased nuclear factor‑κB activity via suppressing the phosphorylation of P65 in lipopolysaccharide (LPS)‑induced human pulmonary alveolar epithelial cells (HPAEpiC) and lung tissues extracted from ALI rats, as determined by immunofluorescence. Furthermore, in vivo experiments demonstrated that the combination of HF and DEX in LPS‑induced ALI rats defended against lung fibrosis, perivascular inflammation, congestion and edema of pulmonary alveoli, as assessed by histopathological analysis, TUNEL staining and immunohistochemistry assay. Taken together, the present study indicated the synergistic effect of HF and DEX on LPS‑induced ALI in HPAEpiC cells and a rat model. These results offer a novel therapeutic approach for the treatment of ALI.
Collapse
Affiliation(s)
- Hai-Lian Du
- Department of Respiratory Medicine, Yidu Central Hospital Affiliated to Weifang Medical College, Qingzhou, Shandong 262500, P.R. China
| | - Ai-Dong Zhai
- Department of Internal Medicine, Maternal and Child Health Hospital of Zibo, Zibo, Shandong 255029, P.R. China
| | - Hong Yu
- Intensive Care Unit, Second Hospital of Harbin City, Harbin, Heilongjiang 150036, P.R. China
| |
Collapse
|
17
|
Abstract
While glucocorticoids have been used for over 50 years to treat rheumatoid and osteoarthritis pain, the prescription of glucocorticoids remains controversial because of potentially harmful side effects at the molecular, cellular and tissue levels. One member of the glucocorticoid family, dexamethasone (DEX) has recently been demonstrated to rescue cartilage matrix loss and chondrocyte viability in animal studies and cartilage explant models of tissue injury and post-traumatic osteoarthritis, suggesting the possibility of DEX as a disease-modifying drug if used appropriately. However, the literature on the effects of DEX on cartilage reveals conflicting results on the drug's safety, depending on the dose and duration of DEX exposure as well as the model system used. Overall, DEX has been shown to protect against arthritis-related changes in cartilage structure and function, including matrix loss, inflammation and cartilage viability. These beneficial effects are not always observed in model systems using initially healthy cartilage or isolated chondrocytes, where many studies have reported significant increases in chondrocyte apoptosis. It is crucially important to understand under what conditions DEX may be beneficial or harmful to cartilage and other joint tissues and to determine potential for safe use of this glucocorticoid in the clinic as a disease-modifying drug.
Collapse
Affiliation(s)
- R. Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A. J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA,Address for correspondence: Prof. Al Grodzinsky, MIT, Centre for Biomedical Engineering, 500 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
18
|
Martín AR, Patel JM, Zlotnick HM, Carey JL, Mauck RL. Emerging therapies for cartilage regeneration in currently excluded 'red knee' populations. NPJ Regen Med 2019; 4:12. [PMID: 31231546 PMCID: PMC6542813 DOI: 10.1038/s41536-019-0074-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 04/29/2019] [Indexed: 12/13/2022] Open
Abstract
The field of articular cartilage repair has made significant advances in recent decades; yet current therapies are generally not evaluated or tested, at the time of pivotal trial, in patients with a variety of common comorbidities. To that end, we systematically reviewed cartilage repair clinical trials to identify common exclusion criteria and reviewed the literature to identify emerging regenerative approaches that are poised to overcome these current exclusion criteria. The term “knee cartilage repair” was searched on clinicaltrials.gov. Of the 60 trials identified on initial search, 33 were further examined to extract exclusion criteria. Criteria excluded by more than half of the trials were identified in order to focus discussion on emerging regenerative strategies that might address these concerns. These criteria included age (<18 or >55 years old), small defects (<1 cm2), large defects (>8 cm2), multiple defect (>2 lesions), BMI >35, meniscectomy (>50%), bilateral knee pathology, ligamentous instability, arthritis, malalignment, prior repair, kissing lesions, neurologic disease of lower extremities, inflammation, infection, endocrine or metabolic disease, drug or alcohol abuse, pregnancy, and history of cancer. Finally, we describe emerging tissue engineering and regenerative approaches that might foster cartilage repair in these challenging environments. The identified criteria exclude a majority of the affected population from treatment, and thus greater focus must be placed on these emerging cartilage regeneration techniques to treat patients with the challenging “red knee”.
Collapse
Affiliation(s)
- Anthony R Martín
- 1McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA.,2Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA
| | - Jay M Patel
- 1McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA.,2Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA
| | - Hannah M Zlotnick
- 1McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA.,2Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA.,3Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - James L Carey
- 1McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Robert L Mauck
- 1McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA.,2Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA.,3Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
19
|
Stefani RM, Halder SS, Estell EG, Lee AJ, Silverstein AM, Sobczak E, Chahine NO, Ateshian GA, Shah RP, Hung CT. A Functional Tissue-Engineered Synovium Model to Study Osteoarthritis Progression and Treatment. Tissue Eng Part A 2019; 25:538-553. [PMID: 30203722 PMCID: PMC6482911 DOI: 10.1089/ten.tea.2018.0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/31/2018] [Indexed: 01/15/2023] Open
Abstract
IMPACT STATEMENT The synovium envelops the diarthrodial joint and plays a key regulatory role in defining the composition of the synovial fluid through filtration and biosynthesis of critical boundary lubricants. Synovium changes often precede cartilage damage in osteoarthritis. We describe a novel in vitro tissue engineered model, validated against native synovium explants, to investigate the structure-function of synovium through quantitative solute transport measures. Synovium was evaluated in the presence of a proinflammatory cytokine, interleukin-1, or the clinically relevant corticosteroid, dexamethasone. We anticipate that a better understanding of synovium transport would support efforts to develop more effective strategies aimed at restoring joint health.
Collapse
Affiliation(s)
- Robert M. Stefani
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Saiti S. Halder
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Eben G. Estell
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Andy J. Lee
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Amy M. Silverstein
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Evie Sobczak
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Nadeen O. Chahine
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Mechanical Engineering, Columbia University, New York, New York
| | - Roshan P. Shah
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
20
|
Kim M, Garrity ST, Steinberg DR, Dodge GR, Mauck RL. Role of dexamethasone in the long-term functional maturation of MSC-laden hyaluronic acid hydrogels for cartilage tissue engineering. J Orthop Res 2018; 36:1717-1727. [PMID: 29178462 PMCID: PMC6948196 DOI: 10.1002/jor.23815] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/23/2017] [Indexed: 02/04/2023]
Abstract
The purpose of study was to investigate the maturation of mesenchymal stem cells (MSC) laden in HA constructs with various combinations of chemically defined medium (CM) components and determine the impact of dexamethasone and serum on construct properties. Constructs were cultured in CM with the addition or withdrawal of media components or were transferred to serum containing media that partially represents an in vivo-like condition where pro-inflammatory signals are present. Constructs cultured in CM+ (CM with TGF-β3) and DEX- (CM+ without dexamethasone) conditions produced robust matrix, while those in ITS/BSA/LA- (CM+ without ITS/BSA/LA) and Serum+ (10% FBS with TGF-β3) produced little matrix. While construct properties in DEX- were greater than those in CM+ at 4 weeks, properties in CM+ and DEX- reversed by 8 weeks. While construct properties in DEX- were greater than those in CM+ at 4 weeks, the continued absence or removal of dexamethasone resulted in marked GAG loss by 8 weeks. Conversely, the continued presence or new addition of dexamethasone at 4 weeks further improved or maintained construct properties through 8 weeks. Finally, when constructs were converted to Serum (in the continued presence of TGF-β3 with or without dexamethasone) after pre-culture in CM+ for 4 weeks, GAG loss was attenuated with addition of dexamethasone. Interestingly, however, collagen content and type was not impacted. In conclusion, dexamethasone influences the functional maturation of MSC-laden HA constructs, and may help to maintain properties during long-term culture or with in vivo translation by repressing pro-inflammatory signals. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1717-1727, 2018.
Collapse
Affiliation(s)
- Minwook Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104,Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, U.S.A
| | - Sean T. Garrity
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David R. Steinberg
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104,Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, U.S.A
| | - George R. Dodge
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104,Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, U.S.A
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104,Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, U.S.A,Address for Correspondence: Robert L. Mauck, Ph.D., Mary Black Ralston Professor of Orthopaedic Surgery, Professor of Bioengineering, McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, Phone: (215) 898-3294, Fax: (215) 573-2133,
| |
Collapse
|
21
|
Dale TP, Forsyth NR. Ectopic Telomerase Expression Fails to Maintain Chondrogenic Capacity in Three-Dimensional Cultures of Clinically Relevant Cell Types. Biores Open Access 2018; 7:10-24. [PMID: 29588876 PMCID: PMC5865620 DOI: 10.1089/biores.2018.0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The poor healing capacity of cartilage and lack of effective treatment for associated disease and trauma makes it a strong candidate for a regenerative medicine approach. Potential therapies tested to date, although effective, have met with a number of intrinsic difficulties possibly related to limited autologous chondrocyte cell yield and quality of cartilage produced. A potential mechanism to bypass limited cell yields and improve quality of differentiation is to immortalize relevant cell types through the ectopic expression of telomerase. Pellet cultures of human chondrocytes (OK3), bone marrow mesenchymal stem cells (BMA13), and embryonic stem cell (H1 line)-derived cells (1C6) and their human telomerase reverse transcriptase (hTERT) transduced counterparts were maintained for 20 days in standard maintenance medium (MM) or transforming growth factor-β3-supplemented prochondrogenic medium (PChM). Pellets were assessed for volume and density by microcomputed tomography. Quantitative gene expression (COL1A2, COL2A1, COL3A1, COL6A3, COL10A1, ACAN, COMP, SOX9); sulfated glycosaminoglycans (sGAGs), and DNA quantification were performed. Histology and immunohistochemistry were used to determine matrix constituent distribution. Pellet culture in PChM resulted in significantly larger pellets with an overall increased density when compared with MM culture. Gene expression analysis revealed similarities in expression patterns between telomerase-transduced and parental cells in both MM and PChM. Of the three parental cell types examined OK3 and BMA13 produced similar amounts of pellet-associated sGAG in PChM (4.62 ± 1.20 and 4.91 ± 1.37 μg, respectively) with lower amounts in 1C6 (2.89 ± 0.52 μg), corresponding to 3.1, 2.3, and 1.6-fold increases from day 0. In comparison, telomerase-transduced cells all had much lower sGAG with OK3H at 2.74 ± 0.11 μg, BMA13H 1.29 ± 0.34 μg, and 1C6H 0.52 ± 0.01 μg corresponding to 1.2, 0.87, and 0.34-fold changes compared with day 0. Histology of day 20 pellets displayed reduced staining overall for collagens and sGAG in telomerase-transduced cells, most notably with alterations in aggrecan and collagen VI; all cells stained positively for collagen II. We conclude that while telomerase transduction may be an effective technique to extend cellular proliferative capacity, it is not sufficient in isolation to sustain a naive chondrogenic phenotype across multiple cell types.
Collapse
Affiliation(s)
- Tina P Dale
- Faculty of Medicine and Health Sciences, Guy Hilton Research Center, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Nicholas R Forsyth
- Faculty of Medicine and Health Sciences, Guy Hilton Research Center, Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
22
|
Liu Y, Li D, Yin Z, Luo X, Liu W, Zhang W, Zhang Z, Cao Y, Liu Y, Zhou G. Prolonged in vitro precultivation alleviates post-implantation inflammation and promotes stable subcutaneous cartilage formation in a goat model. ACTA ACUST UNITED AC 2016; 12:015006. [PMID: 27910822 DOI: 10.1088/1748-605x/12/1/015006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Synthetic biodegradable scaffolds such as polylactic acid coated polyglycolic acid (PLA-PGA) are especially suitable for engineering shaped cartilage such as auricle, but they induce a serious inflammatory reaction particularly in the immunologically aggressive subcutaneous site, leading to resorption of the engineered autologous cartilage. Our previous study in a rabbit model has demonstrated 2 weeks of in vitro precultivation could significantly alleviate the post-implantation inflammation induced by PLA-PGA engineered cartilaginous grafts, but reproduction of this result failed in a preclinical goat model. The aims of the current study were to investigate whether prolonged in vitro precultivation could form a mature cartilaginous graft to resist the acute host response and promote stable subcutaneous cartilage formation in a preclinical goat model. Goat chondrocytes were seeded onto PLA-PGA scaffolds, in vitro precultivated for 2, 4, 8, and 12 weeks, and then implanted subcutaneously in autologous goats for 1 and 8 weeks. The in vitro engineered cartilage (vitro-EC) was examined histologically (hematoxylin and eosin, safranin-O, collagen II). The 1 week explants were examined histologically and stained for CD3, CD68, collagen I, and apoptosis. The 8 week explants were evaluated by histology, wet weight, volume, glycosaminoglycan (GAG) quantification and Young's modulus. With prolonged in vitro time, the quality of vitro-EC improved and the amount of scaffold residue decreased; more pronounced cartilage formation with fewer immune cells (CD3 and CD68 positive), apoptotic cells, and less collagen I expression were observed in explants that had been in vitro precultivated for a longer period. The subcutaneously regenerated neocartilage became more mature after prolonged implantation. These results suggested that prolonged in vitro precultivation allowed formation of a mature cartilaginous graft to resist the acute host response and promoted stable subcutaneous cartilage formation in autologous goats. These findings may provide useful reference for engineering auricle, trachea, nose, and eyelid shaped cartilage, for example.
Collapse
Affiliation(s)
- Yi Liu
- Shanghai 9th People's Hospital, School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University, 639 Zhi Zao Ju Road, Shanghai, People's Republic of China. Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, People's of Republic of China. These authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | |
Collapse
|