1
|
Zhang J, Zhang J, Li H, Zhang H, Meng H. Research progress on biodegradable polymer-based drug delivery systems for the treatment of knee osteoarthritis. Front Bioeng Biotechnol 2025; 13:1561708. [PMID: 40276032 PMCID: PMC12018437 DOI: 10.3389/fbioe.2025.1561708] [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: 01/16/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Knee osteoarthritis (KOA) is a disease that involves multiple anatomical and physiological changes in the knee tissues, including cartilage degeneration, bone remodelling and formation of bony encumbrances, which leads to clinical manifestations including pain, stiffness, swelling and limitation of knee function. Knee osteoarthritis is a chronic joint disease characterised by degenerative cartilage lesions and secondary osteophytes in the knee joint. The symptoms of knee osteoarthritis tend to progress slowly, and at this stage, the number of patients with KOA is increasing. However, due to the adverse effects and poor therapeutic outcomes following surgical treatment, intervention therapy through the utilisation of biodegradable polymeric materials is required. Currently, clinical aspects are mainly used to treat cartilage degeneration in patients with osteoarthritis of the knee by using different kinds of biodegradable biopolymer materials with excellent physical properties, histocompatibility and other properties, combined with a drug delivery system, which can reduce the level of inflammation and stiffness in the focal area, and maximise the restoration of the patient's knee joint joint mobility and athletic ability. Based on the properties of the polymeric material drug delivery system, the polymeric material has a variable drug loading capacity that encapsulates hydrophobic/hydrophilic drugs and controls the release kinetics by regulating the composition and charge. This paper reviews the research progress of Poly (ε-caprolactone) (PCL), Poly(lactic acid) (PLA), Poly (lactic glycolic acid) (PLGA), Poly(ethylene glycol) (PEG) synthetic polymers and collagen, chondroitin sulfate, other natural polymers based drug delivery systems for the treatment of knee osteoarthritis, and explains that different biodegradable polymeric materials have been widely used for the treatment of knee osteoarthritis. However, there are still issues of degradability, toxicity, compatibility, and durability and safety of the drug delivery system of degradable materials that need to be addressed in further clinical trials. As biodegradable biomedical materials continue to be explored, eventually idealized polymeric materials will stand out in the treatment of KOA.
Collapse
Affiliation(s)
- Jinchi Zhang
- Department of Medical, Qingdao Binhai University, Qingdao, China
| | - Jinchao Zhang
- Department of Medical, Qingdao Binhai University, Qingdao, China
| | - Hailong Li
- Department of Medical, Qingdao Binhai University, Qingdao, China
| | - Huimin Zhang
- Department of Nursing, The Third People’s Hospital of Heze, Heze, China
| | - Hongyan Meng
- Department of Medical, Qingdao Binhai University, Qingdao, China
| |
Collapse
|
2
|
Joshi N, Yan J, Dang M, Slaughter K, Wang Y, Wu D, Ung T, Bhingaradiya N, Pandya V, Chen MX, Kaur S, Bhagchandani S, Alfassam HA, Joseph J, Gao J, Dewani M, Chu RWC, Yip RCS, Weldon E, Shah P, Pisal ND, Shukla C, Sherman NE, Luo JN, Conway T, Eickhoff JP, Botelho L, Alhasan AH, Karp JM, Ermann J. A mechanically resilient soft hydrogel improves drug delivery for treating post-traumatic osteoarthritis in physically active joints. Proc Natl Acad Sci U S A 2025; 122:e2409729122. [PMID: 40163719 PMCID: PMC12002200 DOI: 10.1073/pnas.2409729122] [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: 05/17/2024] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Intra-articular delivery of disease-modifying osteoarthritis drugs (DMOADs) is likely to be most effective in the early stages of post-traumatic osteoarthritis (PTOA), when symptoms are minimal, and patients remain physically active. To ensure effective therapy, DMOAD delivery systems therefore must withstand repeated mechanical loading without altering the kinetics of drug release. While soft materials are typically preferred for DMOAD delivery, mechanical loading can compromise their structural integrity and disrupt controlled drug release. In this study, we present a mechanically resilient soft hydrogel that rapidly self-heals under conditions simulating human running while maintaining sustained release of the cathepsin-K inhibitor L-006235, used as a proof-of-concept DMOAD. This hydrogel demonstrated superior performance compared to a previously reported hydrogel designed for intra-articular drug delivery, which, in our study, neither recovered its structure nor maintained drug release under mechanical loading. When injected into mouse knee joints, the hydrogel provided consistent release kinetics of the encapsulated drug in both treadmill-running and nonrunning mice. In a mouse model of severe PTOA exacerbated by treadmill-running, the L-006235 hydrogel significantly reduced cartilage degeneration, whereas the free drug did not. Overall, our data underscore the hydrogel's potential for treating PTOA in physically active patients.
Collapse
Affiliation(s)
- Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Jing Yan
- Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Mickael Dang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Kai Slaughter
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Yufeng Wang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Dana Wu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Trevor Ung
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nutan Bhingaradiya
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Virja Pandya
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Mu Xian Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Shahdeep Kaur
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Sachin Bhagchandani
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Haya A. Alfassam
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
- King Abdulaziz City for Science and Technology Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Jingjing Gao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Mahima Dewani
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Rachel Wai Chun Chu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Ryan Chak Sang Yip
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Eli Weldon
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Purna Shah
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nishkal Dhiraj Pisal
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Chetan Shukla
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nicholas E. Sherman
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - James N. Luo
- Harvard Medical School, Boston, MA02115
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA02115
| | - Thomas Conway
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | | | | | - Ali H. Alhasan
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
- King Abdulaziz City for Science and Technology Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
| | - Jeffrey M. Karp
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Broad Institute, Cambridge, MA02142
- Harvard Stem Cell Institute, Cambridge, MA02138
| | - Joerg Ermann
- Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| |
Collapse
|
3
|
Linderman SW, Barber GF, DeVeaux SA, Botchwey EA, Refai D, Klein AM, García AJ. Dexamethasone Delivery via Amphiphilic, Low-swelling Hydrogels Treats Postoperative Inflammation in Cervical Spine Applications. Adv Healthc Mater 2025; 14:e2404292. [PMID: 39828668 PMCID: PMC11912108 DOI: 10.1002/adhm.202404292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/30/2024] [Indexed: 01/22/2025]
Abstract
Anterior cervical spine surgeries are often complicated by difficulty swallowing due to local postoperative swelling, pain, scarring, and tissue dysfunction. These postoperative events lead to systemic steroid and narcotic use. Local, sustained drug delivery may address these problems, but current materials are unsafe for tight surgical spaces due to high biomaterial swelling, especially upon degradation. To address these shortcomings, a low-swelling, amphiphilic hydrogel system termed DexaPatch is developed containing dexamethasone-poly(lactic-co-glycolic acid) (PLGA) microparticles for sustained release upon local implantation in the surgical site. The bulk amphiphilic hydrogel, comprised of 4-arm poly(ethylene glycol) (PEG)-maleimide macromer cross-linked with triblock dithiolated PEG-poly(propylene glycol)-PEG (poloxamer a.k.a. Pluronic), achieves consistent and tunable mechanical and low-swelling properties. Dexamethasone is released in a burst, followed by a sustained release over 40 days, similar to the release from microparticles alone. The DexaPatch system is lyophilized for shelf stability and surgical handling properties, sterilized, and briefly rehydrated in the operating room prior to surgical implantation in a rabbit model of anterior spinal surgery. DexaPatch results in significantly reduced prevertebral edema radiographically and decreased fibrosis in prevertebral muscles compared to sham surgery. This implantable biomaterial platform reduces local postoperative inflammation with potential surgical applications throughout the body.
Collapse
Affiliation(s)
- Stephen W. Linderman
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332–0363USA
- Emory University School of Medicine100 Woodruff CircleAtlantaGA30322USA
| | - Graham F. Barber
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332–0363USA
| | - S'Dravious A. DeVeaux
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332–0363USA
| | - Edward A. Botchwey
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332–0363USA
| | - Daniel Refai
- Emory University School of Medicine100 Woodruff CircleAtlantaGA30322USA
| | - Adam M. Klein
- Emory University School of Medicine100 Woodruff CircleAtlantaGA30322USA
| | - Andrés J. García
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332–0363USA
| |
Collapse
|
4
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
5
|
Joshi N, Yan J, Dang M, Slaughter K, Wang Y, Wu D, Ung T, Pandya V, Chen MX, Kaur S, Bhagchandani S, Alfassam HA, Joseph J, Gao J, Dewani M, Yip RCS, Weldon E, Shah P, Shukla C, Sherman NE, Luo JN, Conway T, Eickhoff JP, Botelho L, Alhasan AH, Karp JM, Ermann J. A Mechanically Resilient Soft Hydrogel Improves Drug Delivery for Treating Post-Traumatic Osteoarthritis in Physically Active Joints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594611. [PMID: 38826308 PMCID: PMC11142096 DOI: 10.1101/2024.05.16.594611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Intra-articular delivery of disease-modifying osteoarthritis drugs (DMOADs) is likely to be most effective in early post-traumatic osteoarthritis (PTOA) when symptoms are minimal and patients are physically active. DMOAD delivery systems therefore must withstand repeated mechanical loading without affecting the drug release kinetics. Although soft materials are preferred for DMOAD delivery, mechanical loading can compromise their structural integrity and disrupt drug release. Here, we report a mechanically resilient soft hydrogel that rapidly self-heals under conditions resembling human running while maintaining sustained release of the cathepsin-K inhibitor L-006235 used as a proof-of-concept DMOAD. Notably, this hydrogel outperformed a previously reported hydrogel designed for intra-articular drug delivery, used as a control in our study, which neither recovered nor maintained drug release under mechanical loading. Upon injection into mouse knee joints, the hydrogel showed consistent release kinetics of the encapsulated agent in both treadmill-running and non-running mice. In a mouse model of aggressive PTOA exacerbated by treadmill running, L-006235 hydrogel markedly reduced cartilage degeneration. To our knowledge, this is the first hydrogel proven to withstand human running conditions and enable sustained DMOAD delivery in physically active joints, and the first study demonstrating reduced disease progression in a severe PTOA model under rigorous physical activity, highlighting the hydrogel's potential for PTOA treatment in active patients.
Collapse
|
6
|
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
|
7
|
Omidian H, Chowdhury SD. Advancements and Applications of Injectable Hydrogel Composites in Biomedical Research and Therapy. Gels 2023; 9:533. [PMID: 37504412 PMCID: PMC10379998 DOI: 10.3390/gels9070533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Injectable hydrogels have gained popularity for their controlled release, targeted delivery, and enhanced mechanical properties. They hold promise in cardiac regeneration, joint diseases, postoperative analgesia, and ocular disorder treatment. Hydrogels enriched with nano-hydroxyapatite show potential in bone regeneration, addressing challenges of bone defects, osteoporosis, and tumor-associated regeneration. In wound management and cancer therapy, they enable controlled release, accelerated wound closure, and targeted drug delivery. Injectable hydrogels also find applications in ischemic brain injury, tissue regeneration, cardiovascular diseases, and personalized cancer immunotherapy. This manuscript highlights the versatility and potential of injectable hydrogel nanocomposites in biomedical research. Moreover, it includes a perspective section that explores future prospects, emphasizes interdisciplinary collaboration, and underscores the promising future potential of injectable hydrogel nanocomposites in biomedical research and applications.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Sumana Dey Chowdhury
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
8
|
Zhang X, Wang Z, Wang B, Li J, Yuan H. lncRNA OIP5-AS1 attenuates the osteoarthritis progression in IL-1β-stimulated chondrocytes. Open Med (Wars) 2023; 18:20230721. [PMID: 37333451 PMCID: PMC10276615 DOI: 10.1515/med-2023-0721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 04/18/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
In view of the association between long noncoding RNA OIP5-AS1 and osteoarthritis (OA) pathology, the corresponding potential mechanism is worthy of exploration. Primary chondrocytes were identified by morphological observation and immunohistochemical staining of collagen II. The association between OIP5-AS1 and miR-338-3p was analyzed by StarBase and dual-luciferase reporter assay. After the expression of OIP5-AS1 or miR-338-3p in interleukin (IL)-1β-stimulated primary chondrocytes and CHON-001 cells was manipulated, cell viability, proliferation, apoptosis rate, apoptosis-related protein (cleaved caspase-9, Bax) expressions, extracellular matrix (ECM) (matrix metalloproteinase (MMP)-3, MMP-13, aggrecan, and collagen II), PI3K/AKT pathway, and mRNA expressions of inflammatory factors (IL-6 and IL-8), OIP5-AS1, and miR-338-3p were determined by cell counting kit-8, EdU, flow cytometry, Western blot, and quantitative reverse transcription-polymerase chain reaction. As a result, the expression of OIP5-AS1 was downregulated in IL-1β-activated chondrocytes, while miR-338-3p was overexpressed. OIP5-AS1 overexpression reversed the effects of IL-1β on viability, proliferation, apoptosis, ECM degradation, and inflammation in chondrocytes. However, OIP5-AS1 knockdown exhibited opposite effects. Interestingly, the effects of OIP5-AS1 overexpression were partially offset by miR-338-3p overexpression. Furthermore, OIP5-AS1 overexpression blocked the PI3K/AKT pathway by modulating miR-338-3p expression. In sum, OIP5-AS1 promotes viability and proliferation, and inhibits apoptosis and ECM degradation in IL-1β-activated chondrocytes by targeting miR-338-3p through blocking the PI3K/AKT pathway, indicating an attractive strategy for OA treatment.
Collapse
Affiliation(s)
- Xuefeng Zhang
- The First Clinical Medical College, Jinan University, Guangzhou, Guangdong 510632, P.R. China
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Zhikun Wang
- Department of Orthopedics, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Binbin Wang
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Jingyi Li
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Hui Yuan
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| |
Collapse
|
9
|
Delbaldo C, Tschon M, Martini L, Fini M, Codispoti G. Benefits of Applying Nanotechnologies to Hydrogels in Efficacy Tests in Osteoarthritis Models-A Systematic Review of Preclinical Studies. Int J Mol Sci 2022; 23:ijms23158236. [PMID: 35897805 PMCID: PMC9368605 DOI: 10.3390/ijms23158236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 12/09/2022] Open
Abstract
Osteoarthritis (OA) is a severe musculoskeletal disease with an increasing incidence in the worldwide population. Recent research has focused on the development of innovative strategies to prevent articular cartilage damage and slow down OA progression, and nanotechnologies applied to hydrogels have gained particular interest. The aim of this systematic review is to investigate the state of the art on preclinical in vitro and in vivo efficacy studies applying nanotechnologies to hydrogels in OA models to elucidate the benefits of their applications. Three databases were consulted for eligible papers. The inclusion criteria were in vitro and in vivo preclinical studies, using OA cells or OA animal models, and testing hydrogels and nanoparticles (NPs) over the last ten years. Data extraction and quality assessment were performed. Eleven papers were included. In vitro studies evidenced that NP-gels do not impact on cell viability and do not cause inflammation in OA cell phenotypes. In vivo research on rodents showed that these treatments could increase drug retention in joints, reducing inflammation and preventing articular cartilage damage. Nanotechnologies in preclinical efficacy tests are still new and require extensive studies and technical hits to determine the efficacy, safety, fate, and localization of NPs for translation into an effective therapy for OA patients.
Collapse
|
10
|
Advances in nanoenabled 3D matrices for cartilage repair. Acta Biomater 2022; 150:1-21. [PMID: 35902038 DOI: 10.1016/j.actbio.2022.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
Cartilage repair strategies are evolving at a fast pace with technology development. Matrices that offer multifaceted functions and a full adaption to the cartilage defect are of pivotal interest. Current cartilage repair strategies face numerous challenges, mostly related to the development of highly biomimetic materials, non-invasive injectable solutions, and adequate degradation rates. These strategies often fail due to feeble mechanical properties, the inability to sustain cell adhesion, growth, and differentiation or by underestimating other players of cartilage degeneration, such as the installed pro-inflammatory microenvironment. The integration of nanomaterials (NMs) into 3D scaffolds, hydrogels and bioinks hold great potential in the improvement of key features of materials that are currently applied in cartilage tissue engineering strategies. NMs offer a high surface to volume ratio and their multiple applications can be explored to enhance cartilage mechanical properties, biocompatibility, cell differentiation, inflammation modulation, infection prevention and even to function as diagnostic tools or as stimuli-responsive cues in these 3D structures. In this review, we have critically reviewed the latest advances in the development of nanoenabled 3D matrices - enhanced by means of NMs - in the context of cartilage regeneration. We have provided a wide perspective of the synergistic effect of combining 3D strategies with NMs, with emphasis on the benefits brought by NMs in achieving functional and enhanced therapeutic outcomes. STATEMENT OF SIGNIFICANCE: Cartilage is one of the most challenging tissues to treat owing to its limited self-regeneration potential. Novel strategies using nanoenabled 3D matrices have emerged from the need to design more efficient solutions for cartilage repair, that take into consideration its unique mechanical properties and can direct specific cell behaviours. Here we aim to provide a comprehensive review on the synergistic effects of 3D matrices nanoenrichment in the context of cartilage regeneration, with emphasis on the heightening brought by nanomaterials in achieving functional and enhanced therapeutic outcomes. We anticipate this review to provide a wide perspective on the past years' research on the field, demonstrating the great potential of these approaches in the treatment and diagnosis of cartilage-related disorders.
Collapse
|
11
|
Yi X, Xu Z, Liu Q, Zhou H, Yuan L, Li D, Zhao L, Mu C, Ge L. Matrix metalloproteinase-responsive collagen-oxidized hyaluronic acid injectable hydrogels for osteoarthritic therapy. BIOMATERIALS ADVANCES 2022; 137:212804. [PMID: 35929283 DOI: 10.1016/j.bioadv.2022.212804] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
Drug delivery system and intra-articular injection have been clinically applied to prolong drug residence time and reduce side effects in the treatment of osteoarthrosis. Herein, injectable hydrogels with sustained-dexamethasone sodium phosphate (DSP) release behavior in response to matrix metalloproteinase (MMP) were developed for osteoarthritic therapy. Hyaluronic acid undergoes specific oxidation in the present of sodium periodate to prepare oxidized hyaluronic acid (OHA). Then the DSP-loaded collagen-based hydrogels (Col-OHA) were developed by the Schiff's base crosslinking between OHA and Type I collagen besides the self-assembly of collagen induced by OHA. The results indicate that the collagen self-assembly into collagen fibrils makes great contribution for shortening gelation time of Col-OHA hydrogels. Col-OHA hydrogels possess interconnected porous microstructure, good injectability, excellent self-healing performance, strong mechanical property, low swelling ability, good blood compatibility and no cytotoxicity. Significantly, Col-OHA hydrogels show highly sensitive and significantly substantially sustained release of DSP in response to MMP. DSP-loaded Col-OHA hydrogel possesses significant inhibition for the production of inflammatory cytokines in the joint synovium, which can effectively relieve the symptoms of osteoarthritis continuously. Col-OHA hydrogel has no obvious effect on liver and kidney functions. Overall, the Col-OHA hydrogels with excellent biocompatibility are the promising drug-loading system for the intra-articular injection therapy of osteoarthrosis.
Collapse
Affiliation(s)
- Xueling Yi
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Zhilang Xu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Qisong Liu
- Research Institute of Natural Gas Technology, Petro China Southwest Oil & Gas Field Company, Chengdu 610213, PR China
| | - Hongmei Zhou
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Lun Yuan
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Defu Li
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Lei Zhao
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, 610041, PR China
| | - Changdao Mu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Liming Ge
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China.
| |
Collapse
|
12
|
Di Francesco M, Fragassi A, Pannuzzo M, Ferreira M, Brahmachari S, Decuzzi P. Management of osteoarthritis: From drug molecules to nano/micromedicines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1780. [PMID: 35253405 PMCID: PMC9285805 DOI: 10.1002/wnan.1780] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
Abstract
With the change in lifestyle and aging of the population, osteoarthritis (OA) is emerging as a major medical burden globally. OA is a chronic inflammatory and degenerative disease initially manifesting with joint pain and eventually leading to permanent disability. To date, there are no drugs available for the definitive treatment of osteoarthritis and most therapies have been palliative in nature by alleviating symptoms rather than curing the disease. This coupled with the vague understanding of the early symptoms and methods of diagnosis so that the disease continues as a global problem and calls for concerted research efforts. A cascade of events regulates the onset and progression of osteoarthritis starting with the production of proinflammatory cytokines, including interleukin (IL)‐1β, IL‐6, tumor necrosis factor (TNF)‐α; catabolic enzymes, such as matrix metalloproteinases (MMPs)‐1, ‐3, and ‐13, culminating into cartilage breakdown, loss of lubrication, pain, and inability to load the joint. Although intra‐articular injections of small and macromolecules are often prescribed to alleviate symptoms, low residence times within the synovial cavity severely impair their efficacy. This review will briefly describe the factors dictating the onset and progression of the disease, present the current clinically approved methods for its treatment and diagnosis, and finally elaborate on the main challenges and opportunities for the application of nano/micromedicines in the treatment of osteoarthritis. Thus, future treatment regimens will benefit from simultaneous consideration of the mechanobiological, the inflammatory, and tissue degradation aspects of the disease. This article is categorized under:Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Agnese Fragassi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Chemistry and Industrial Chemistry, University of Genova, Genoa, Italy
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
13
|
Gan Q, Pan H, Zhang W, Yuan Y, Qian J, Liu C. Fabrication and evaluation of a BMP-2/dexamethasone co-loaded gelatin sponge scaffold for rapid bone regeneration. Regen Biomater 2022; 9:rbac008. [PMID: 35592142 PMCID: PMC9113239 DOI: 10.1093/rb/rbac008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 11/26/2022] Open
Abstract
Improving the osteogenic activity of BMP-2 in vivo has significant clinical application value. In this research, we use a clinical gelatin sponge scaffold loaded with BMP-2 and dexamethasone (Dex) to evaluate the osteogenic activity of dual drugs via ectopic osteogenesis in vivo. We also investigate the mechanism of osteogenesis induced by BMP-2 and Dex with C2C12, a multipotent muscle-derived progenitor cell. The results show that the gelatin scaffold with Dex and BMP-2 can significantly accelerate osteogenesis in vivo. It is indicated that compared with the BMP-2 or Dex alone, 100 nM of Dex can dramatically enhance the BMP-2-induced alkaline phosphatase activity (ALP), ALP mRNA expression and mineralization. Further studies show that 100 nM of Dex can maintain the secondary structure of BMP-2 and facilitate recognition of BMP-2 with its receptors on the surface of C2C12 cells. We also find that in C2C12, Dex has no obvious effect on the BMP-2-induced Smad1/5/8 protein expression and the STAT3-dependent pathway, but Runx2-dependent pathway is involved in the Dex-stimulated osteoblast differentiation of BMP-2 both in vitro and in vivo. Based on these results, a potential mechanism model about the synergistic osteoinductive effect of Dex and BMP-2 in C2C12 cells via Runx2 activation is proposed. This may provide a theoretical basis for the pre-clinical application of Dex and BMP-2 for bone regeneration.
Collapse
Affiliation(s)
- Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Hao Pan
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Wenjing Zhang
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jiangchao Qian
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
14
|
Chilbule SK, Rajagopal K, Walter N, Dutt V, Madhuri V. Role of WNT Agonists, BMP and VEGF Antagonists in Rescuing Osteoarthritic Knee Cartilage in a Rat Model. Indian J Orthop 2022; 56:24-33. [PMID: 35070139 PMCID: PMC8748585 DOI: 10.1007/s43465-021-00434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/27/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The superficial zone of articular cartilage (AC) is vital for its function and biomechanics. The damaged AC gets vascularized and undergoes hypertrophy and ossification. Studies have highlighted these two as the major causative factors in osteoarthritis (OA). We aimed at preventing the OA progression in a rat knee instability model by inhibiting the vascular ingrowth and ossification using VEGF and BMP antagonist. A WNT agonist was also used to promote AC regeneration because of its protective effect on the superficial layer. METHODS Rat knee OA was created by surgical excision of the medial meniscus and medial collateral ligament. Forty rats were divided into two groups of twenty each for surgical control and tests (surgery + intra-articular injection of drugs every two weeks). Ten animals from each group were sacrificed at four and eight weeks. Histology was mainly used to evaluate the outcome. RESULTS A surgical OA model was successfully created with higher histological scores for operated knees, both in short- (P = 0.0001) and long-term (P = 0.001). Modified Mankin score was lesser in the test animals as compared to control (P = 0.17) in the short-term, but the trend was reversed in the long-term (P = 0.13). Subgroup analysis revealed that repeated injections in the anterolateral compartment contributed to higher scores in the lateral (P = 0.03) and anterior (P = 0.03) compartment of the knee in the long-term. CONCLUSION The combinatorial approach was effective in controlling the OA in short-term. Further studies are needed to test the sustained drug delivery system to improve the outcome.
Collapse
Affiliation(s)
- Sanjay K. Chilbule
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
| | - Karthikeyan Rajagopal
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
- Centre for Stem Cell Research, Christian Medical College, Vellore, 632002 India
| | - Noel Walter
- Department of Forensic Medicine, Christian Medical College, Vellore, 632004 India
| | - Vivek Dutt
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
| | - Vrisha Madhuri
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
- Centre for Stem Cell Research, Christian Medical College, Vellore, 632002 India
| |
Collapse
|
15
|
Klimak M, Nims RJ, Pferdehirt L, Collins KH, Harasymowicz NS, Oswald SJ, Setton LA, Guilak F. Immunoengineering the next generation of arthritis therapies. Acta Biomater 2021; 133:74-86. [PMID: 33823324 DOI: 10.1016/j.actbio.2021.03.062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022]
Abstract
Immunoengineering continues to revolutionize healthcare, generating new approaches for treating previously intractable diseases, particularly in regard to cancer immunotherapy. In joint diseases, such as osteoarthritis (OA) and rheumatoid arthritis (RA), biomaterials and anti-cytokine treatments have previously been at that forefront of therapeutic innovation. However, while many of the existing anti-cytokine treatments are successful for a subset of patients, these treatments can also pose severe risks, adverse events and off-target effects due to continuous delivery at high dosages or a lack of disease-specific targets. The inadequacy of these current treatments has motivated the development of new immunoengineering strategies that offer safer and more efficacious alternative therapies through the precise and controlled targeting of specific upstream immune responses, including direct and mechanistically-driven immunoengineering approaches. Advances in the understanding of the immunomodulatory pathways involved in musculoskeletal disease, in combination with the growing emphasis on personalized medicine, stress the need for carefully considering the delivery strategies and therapeutic targets when designing therapeutics to better treat RA and OA. Here, we focus on recent advances in biomaterial and cell-based immunomodulation, in combination with genetic engineering, for therapeutic applications in joint diseases. The application of immunoengineering principles to the study of joint disease will not only help to elucidate the mechanisms of disease pathogenesis but will also generate novel disease-specific therapeutics by harnessing cellular and biomaterial responses. STATEMENT OF SIGNIFICANCE: It is now apparent that joint diseases such as osteoarthritis and rheumatoid arthritis involve the immune system at both local (i.e., within the joint) and systemic levels. In this regard, targeting the immune system using both biomaterial-based or cellular approaches may generate new joint-specific treatment strategies that are well-controlled, safe, and efficacious. In this review, we focus on recent advances in immunoengineering that leverage biomaterials and/or genetically engineered cells for therapeutic applications in joint diseases. The application of such approaches, especially synergistic strategies that target multiple immunoregulatory pathways, has the potential to revolutionize our understanding, treatment, and prevention of joint diseases.
Collapse
Affiliation(s)
- Molly Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Robert J Nims
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Sara J Oswald
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Lori A Setton
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
16
|
Han X, Wu Y, Shan Y, Zhang X, Liao J. Effect of Micro-/Nanoparticle Hybrid Hydrogel Platform on the Treatment of Articular Cartilage-Related Diseases. Gels 2021; 7:gels7040155. [PMID: 34698122 PMCID: PMC8544595 DOI: 10.3390/gels7040155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
Joint diseases that mainly lead to articular cartilage injury with prolonged severe pain as well as dysfunction have remained unexplained for many years. One of the main reasons is that damaged articular cartilage is unable to repair and regenerate by itself. Furthermore, current therapy, including drug therapy and operative treatment, cannot solve the problem. Fortunately, the micro-/nanoparticle hybrid hydrogel platform provides a new strategy for the treatment of articular cartilage-related diseases, owing to its outstanding biocompatibility, high loading capability, and controlled release effect. The hybrid platform is effective for controlling symptoms of pain, inflammation and dysfunction, and cartilage repair and regeneration. In this review, we attempt to summarize recent studies on the latest development of micro-/nanoparticle hybrid hydrogel for the treatment of articular cartilage-related diseases. Furthermore, some prospects are proposed, aiming to improve the properties of the micro-/nanoparticle hybrid hydrogel platform so as to offer useful new ideas for the effective and accurate treatment of articular cartilage-related diseases.
Collapse
|
17
|
Paredes J, Pekmezian A, Andarawis-Puri N. MRL/MpJ tendon matrix-derived therapeutic promotes improved healing outcomes in scar-mediated canonical tendon healing. J Orthop Res 2021; 39:1548-1560. [PMID: 32441819 PMCID: PMC7680300 DOI: 10.1002/jor.24754] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/10/2020] [Accepted: 05/08/2020] [Indexed: 02/04/2023]
Abstract
Tendons are commonly injured connective soft tissues characterized by an ineffective healing response that results in scar formation and loss of functional and structural properties. Naturally occurring extracellular matrix (ECM) constructs have become a promising therapeutic for tendon injuries due to their capacity to harness a complex biological environment. However, in tendon, the ECM properties needed for improved healing remain unknown. Interestingly, we have determined that the improved tendon healing response of the scarless-healing MRL/MpJ is driven by intrinsic properties with therapeutic potential to modulate the proliferative and morphological behavior of cells derived from a canonically healing model in vitro. We hypothesize that a distinct composition of ECM deposited during the early healing response of the MRL/MpJ will harnesses the biological cues to stimulate improved structure and function in vivo of canonically healing B6 mice. Accordingly, MRL/MpJ and B6 patellar tendons were injured via midsubstance punch defects. Healing tendons were isolated after 3 or 7 days and encapsulated in PEG-4MAL hydrogels to develop ECM-derived therapeutic constructs. Constructs were then introduced into B6 mice as a treatment following full thickness midsubstance-punch injuries. Treatment with ECM-derived constructs from MRL/MpJ tendons after 7-days post-injury (M7) resulted in improved matrix alignment, tissue stiffness, decreased collagen III content and improved cell morphology in B6 tendons after 6 weeks post-injury. Furthermore, proteomic analysis showed that M7 contained a unique compositional profile rich in glycoproteins, thereby elucidating a valuable naturally-derived platform for the treatment of tendon injuries. Overall this work highlights promising targets for future therapeutic development and tissue engineering applications.
Collapse
Affiliation(s)
- Juan Paredes
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Ashley Pekmezian
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Nelly Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York,Hospital for Special Surgery, New York, New York
| |
Collapse
|
18
|
Mason D, Englund M, Watt FE. Prevention of posttraumatic osteoarthritis at the time of injury: Where are we now, and where are we going? J Orthop Res 2021; 39:1152-1163. [PMID: 33458863 DOI: 10.1002/jor.24982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/11/2021] [Indexed: 02/04/2023]
Abstract
This overview of progress made in preventing post-traumatic osteoarthritis (PTOA) was delivered in a workshop at the Orthopaedics Research Society Annual Conference in 2019. As joint trauma is a major risk factor for OA, defining the molecular changes within the joint at the time of injury may enable the targeting of biological processes to prevent later disease. Animal models have been used to test therapeutic targets to prevent PTOA. A review of drug treatments for PTOA in rodents and rabbits between 2016 and 2018 revealed 11 systemic interventions, 5 repeated intra-articular or topical interventions, and 5 short-term intra-articular interventions, which reduced total Osteoarthritis Research Society International scores by 30%-50%, 20%-70%, and 0%-40%, respectively. Standardized study design, reporting of effect size, and quality metrics, alongside a "whole joint" approach to assessing efficacy, would improve the translation of promising new drugs. A roadblock to translating preclinical discoveries has been the lack of guidelines on the design and conduct of human trials to prevent PTOA. An international workshop addressing this in 2016 considered inclusion criteria and study design, and advocated the use of experimental medicine studies to triage candidate treatments and the development of early biological and imaging biomarkers. Human trials for the prevention of PTOA have tested anakinra after anterior cruciate ligament rupture and dexamethasone after radiocarpal injury. PTOA offers a unique opportunity for defining early mechanisms of OA to target therapeutically. Progress in trial design and high-quality preclinical research, and allegiance with patients, regulatory bodies, and the pharmaceutical industry, will advance this field.
Collapse
Affiliation(s)
- Deborah Mason
- Biomechanics and Bioengineeering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Martin Englund
- Faculty of Medicine, Department of Clinical Sciences Lund, Orthopedics, Clinical Epidemiology Unit, Lund Unversity, Lund, Sweden
| | - Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Berke IM, Jain E, Yavuz B, McGrath T, Chen L, Silva MJ, Mbalaviele G, Guilak F, Kaplan DL, Setton LA. NF-κB-mediated effects on behavior and cartilage pathology in a non-invasive loading model of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2021; 29:248-256. [PMID: 33246158 PMCID: PMC8023431 DOI: 10.1016/j.joca.2020.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study aimed to examine the temporal activation of NF-κB and its relationship to the development of pain-related sensitivity and behavioral changes in a non-invasive murine knee loading model of PTOA. METHOD Following knee injury NF-κB activity was assessed longitudinally via in vivo imaging in FVB. Cg-Tg (HIV-EGFP,luc)8Tsb/J mice. Measures of pain-related sensitivity and behavior were also assessed longitudinally for 16 weeks. Additionally, we antagonized NF-κB signaling via intra-articular delivery of an IκB kinase two antagonist to understand how local NF-κB inhibition might alter disease progression. RESULTS Following joint injury NF-κB signaling within the knee joint was transiently increased and peaked on day 3 with an estimated 1.35 p/s/cm2/sr (95% CI 0.913.1.792 p/s/cm2/sr) fold increase in signaling when compared to control joints. Furthermore, injury resulted in the long-term development of hindpaw allodynia. Hyperalgesia withdrawal thresholds were reduced at injured knee joints, with the largest reduction occurring 2 days following injury (estimate of between group difference 129.1 g with 95% CI 60.9,197.4 g), static weight bearing on injured limbs was also reduced. Local delivery of an NF-κB inhibitor following joint injury reduced chondrocyte death and influenced the development of pain-related sensitivity but did not reduce long-term cartilage degeneration. CONCLUSION These findings underscore the development of behavioral changes in this non-invasive loading model of PTOA and their relationships to NF-κB activation and pathology. They also highlight the potential chondroprotective effects of NF-κB inhibition shortly following joint injury despite limitations in preventing the long-term development of joint degeneration in this model of PTOA.
Collapse
Affiliation(s)
- I M Berke
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - E Jain
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - B Yavuz
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, 02155, USA
| | - T McGrath
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - L Chen
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - M J Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - G Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - F Guilak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
| | - D L Kaplan
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, 02155, USA
| | - L A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
20
|
DeJulius CR, Gulati S, Hasty KA, Crofford LJ, Duvall CL. Recent Advances in Clinical Translation of Intra-Articular Osteoarthritis Drug Delivery Systems. ADVANCED THERAPEUTICS 2021; 4:2000088. [PMID: 33709019 PMCID: PMC7941755 DOI: 10.1002/adtp.202000088] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) is a degenerative disease of the joints and a leading cause of physical disability in adults. Intra-articular (IA) therapy is a popular treatment strategy for localized, single-joint OA; however, small-molecule drugs such as corticosteroids do not provide prolonged relief. One possible reason for their lack of efficacy is high clearance rates from the joint through constant lymphatic drainage of the synovial tissues and synovial fluid and also by their exchange via the synovial vasculature. Advanced drug delivery strategies for extended release of therapeutic agents in the joint space is a promising approach to improve outcomes for OA patients. Broadly, the basic principle behind this strategy is to encapsulate therapeutic agents in a polymeric drug delivery system (DDS) for diffusion- and/or degradation-controlled release, whereby degradation can occur by hydrolysis or tied to relevant microenvironmental cues such as pH, reactive oxygen species (ROS), and protease activity. In this review, we highlight the development of clinically tested IA therapies for OA and highlight recent systems which have been investigated preclinically. DDS strategies including hydrogels, liposomes, polymeric microparticles (MPs) and nanoparticles (NPs), drug conjugates, and combination systems are introduced and evaluated for clinical translational potential.
Collapse
Affiliation(s)
- Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37232, United States
| | - Shubham Gulati
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37232, United States
| | - Karen A Hasty
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, 1211 Union Ave. Suite 520, Memphis, TN 38104, United States
| | - Leslie J Crofford
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, 1161 21 Ave. S., Nashville, TN 37232, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37232, United States
| |
Collapse
|
21
|
Mancipe Castro LM, Sequeira A, García AJ, Guldberg RE. Articular Cartilage- and Synoviocyte-Binding Poly(ethylene glycol) Nanocomposite Microgels as Intra-Articular Drug Delivery Vehicles for the Treatment of Osteoarthritis. ACS Biomater Sci Eng 2020; 6:5084-5095. [PMID: 33455260 PMCID: PMC8221079 DOI: 10.1021/acsbiomaterials.0c00960] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Intra-articular (IA) injection is an attractive route of administration for the treatment of osteoarthritis (OA). However, free drugs injected into the joint space are rapidly cleared and many of them can induce adverse off-target effects on different IA tissues. To overcome these limitations, we designed nanocomposite 4-arm-poly(ethylene glycol)-maleimide (PEG-4MAL) microgels, presenting cartilage- or synoviocyte-binding peptides, containing poly(lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) as an IA small molecule drug delivery system. Microgels containing rhodamine B (model drug)-loaded PLGA NPs were synthesized using microfluidics technology and exhibited a sustained, near zero-order release of the fluorophore over 16 days in vitro. PEG-4MAL microgels presenting synoviocyte- or cartilage-targeting peptides specifically bound to rabbit and human synoviocytes or to bovine articular cartilage in vitro, respectively. Finally, using a rat model of post-traumatic knee OA, PEG-4MAL microgels were shown to be retained in the joint space for at least 3 weeks without inducing any joint degenerative changes as measured by EPIC-μCT and histology. Additionally, all microgel formulations were found trapped in the synovial membrane and significantly increased the IA retention time of a model small molecule near-infrared (NIR) dye compared to that of the free dye. These results suggest that peptide-functionalized nanocomposite PEG-4MAL microgels represent a promising intra-articular vehicle for tissue-localized drug delivery and prolonged IA drug retention for the treatment of OA.
Collapse
Affiliation(s)
- Lina María Mancipe Castro
- Parker H. Petit Institute for Bioengineering and
Biosciences, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332,
U.S.A
- George W. Woodruff School of Mechanical Engineering,
Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, U.S.A
| | - Abigail Sequeira
- School of Chemical and Biomolecular Engineering, Georgia
Institute of Technology, 311 Ferst Drive NW, Atlanta, GA 30332, U.S.A
| | - Andrés J. García
- Parker H. Petit Institute for Bioengineering and
Biosciences, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332,
U.S.A
- George W. Woodruff School of Mechanical Engineering,
Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, U.S.A
| | - Robert E. Guldberg
- Phil and Penny Knight Campus for Accelerating Scientific
Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403-6231
| |
Collapse
|
22
|
Therapeutic Manipulation of Macrophages Using Nanotechnological Approaches for the Treatment of Osteoarthritis. NANOMATERIALS 2020; 10:nano10081562. [PMID: 32784839 PMCID: PMC7466380 DOI: 10.3390/nano10081562] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is the most common joint pathology causing severe pain and disability. Macrophages play a central role in the pathogenesis of OA. In the joint microenvironment, macrophages with an M1-like pro-inflammatory phenotype induce chronic inflammation and joint destruction, and they have been correlated with the development and progression of the disease, while the M2-like anti-inflammatory macrophages support the recovery of the disease, promoting tissue repair and the resolution of inflammation. Nowadays, the treatment of OA in the clinic relies on systemic and/or intra-articular administration of anti-inflammatory and pain relief drugs, as well as surgical interventions for the severe cases (i.e., meniscectomy). The disadvantages of the pharmacological therapy are related to the chronic nature of the disease, requiring prolonged treatments, and to the particular location of the pathology in joint tissues, which are separated anatomical compartments with difficult access for the drugs. To overcome these challenges, nanotechnological approaches have been investigated to improve the delivery of drugs toward macrophages into the diseased joint. This strategy may offer advantages by reducing off-target toxicities and improving long-term therapeutic efficacy. In this review, we describe the nanomaterial-based approaches designed so far to directly or indirectly manipulate macrophages for the treatment of osteoarthritis.
Collapse
|