1
|
Morici L, Allémann E, Rodríguez-Nogales C, Jordan O. Cartilage-targeted drug nanocarriers for osteoarthritis therapy. Int J Pharm 2024; 666:124843. [PMID: 39424088 DOI: 10.1016/j.ijpharm.2024.124843] [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: 07/30/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Osteoarthritis (OA) is a joint disease common worldwide. Currently, no disease-modifying osteoarthritis drugs (DMOADs) have successfully passed clinical trials, often due to a lack of cartilage penetration. Thus, targeting the extracellular matrix (ECM) is a major priority. The design of cartilage-targeting drug delivery systems (DDSs) for intra-articular administration requires consideration of the physicochemical properties of articular cartilage, such as its porosity and negative fixed charge. Various positively charged biomaterials such as polyaminoacids, proteins, polymers, and lipids can be used as DDSs to enhance cartilage penetration. Cationic nanocarriers interact electrostatically with anionic glycosaminoglycans of the ECM, ensuring passive cartilage-targeting penetration and prolonged retention. Active targeting strategies involve DDSs surface decoration using antibodies or peptides with a strong affinity for collagen II and chondrocytes in the cartilage. This review presents all the relevant bio-physicochemical properties of healthy and OA cartilages, as well as state-of-the-art intra-articular cartilage-targeted DDSs, intending to better understand the recent advances in the application of cartilage-targeting delivery systems for OA therapy.
Collapse
Affiliation(s)
- Luca Morici
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Carlos Rodríguez-Nogales
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland.
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland.
| |
Collapse
|
2
|
Carter BM, Day GJ, Zhang WH, Sessions RB, Jackson CJ, Perriman AW. Partitioning of an Enzyme-Polymer Surfactant Nanocomplex into Lipid-Rich Cellular Compartments Drives In Situ Hydrolysis of Organophosphates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2401982. [PMID: 38992997 DOI: 10.1002/smll.202401982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/30/2024] [Indexed: 07/13/2024]
Abstract
Most organophosphates (OPs) are hydrophobic, and after exposure, can sequester into lipophilic regions within the body, such as adipose tissue, resulting in long term chronic effects. Consequently, there is an urgent need for therapeutic agents that can decontaminate OPs in these hydrophobic regions. Accordingly, an enzyme-polymer surfactant nanocomplex is designed and tested comprising chemically supercharged phosphotriesterase (Agrobacterium radiobacter; arPTE) electrostatically conjugated to amphiphilic polymer surfactant chains ([cat.arPTE][S-]). Experimentally-derived structural data are combined with molecular dynamics (MD) simulations to provide atomic level detail on conformational ensembles of the nanocomplex using dielectric constants relevant to aqueous and lipidic microenvironments. These show the formation of a compact admicelle pseudophase surfactant corona under aqueous conditions, which reconfigures to yield an extended conformation at a low dielectric constant, providing insight into the mechanism underpinning cell membrane binding. Significantly, it demonstrated that [cat.arPTE][S-] spontaneously binds to human mesenchymal stem cell membranes (hMSCs), resulting in on-cell OP hydrolysis. Moreover, the nanoconstruct can endocytose and partition into the intracellular fatty vacuoles of adipocytes and hydrolyze sequestered OP.
Collapse
Affiliation(s)
- Benjamin M Carter
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Graham J Day
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - William H Zhang
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Colin J Jackson
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, ACT, 2601, Australia
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
3
|
Nhan J, Strebel N, Virah Sawmy K, Yin J, St-Pierre JP. Characterization of Calcium- and Strontium-Polyphosphate Particles Toward Drug Delivery into Articular Cartilage. Macromol Biosci 2024; 24:e2300345. [PMID: 37777870 DOI: 10.1002/mabi.202300345] [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: 07/27/2023] [Revised: 09/21/2023] [Indexed: 10/02/2023]
Abstract
Drug delivery into articular cartilage poses many challenges due in part to its lack of vasculature. While intra-articular injections are effective for the local administration of drugs, small molecules are rapidly cleared from the synovial fluid. As such, there is a need to develop effective drug delivery strategies to improve the residence times of bioactive molecules in the joint and elicit a sustained therapeutic effect. In this study, calcium- and strontium-polyphosphate particles are synthesized and characterized as potential drug carriers into articular cartilage. Physicochemical characterization reveals that the particles exhibit a spherical morphology, have a negative zeta potential, and are nanoscale in size. Biological characterization in chondrocytes confirms cellular uptake of the particles and demonstrates both size and concentration-dependent cytotoxicity at high concentrations. Furthermore, treatment of chondrocytes with these particles results in a reduction in cell proliferation and metabolic activity, confirming biological effects. Finally, incubation with cartilage tissue explants suggests successful uptake, despite the particles exhibiting a negative surface charge. Therefore, from the results of this study, these polyphosphate-based particles have potential as a drug carrier into articular cartilage and warrant further development.
Collapse
Affiliation(s)
- Jordan Nhan
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Nicolas Strebel
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Khushnouma Virah Sawmy
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Jordan Yin
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| |
Collapse
|
4
|
Gong J, Nhan J, St-Pierre JP, Gillies ER. Designing polymers for cartilage uptake: effects of architecture and molar mass. J Mater Chem B 2023; 11:8804-8816. [PMID: 37668597 DOI: 10.1039/d3tb01417g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Osteoarthritis (OA) is a progressive disease, involving the progressive breakdown of cartilage, as well as changes to the synovium and bone. There are currently no disease-modifying treatments available clinically. An increasing understanding of the disease pathophysiology is leading to new potential therapeutics, but improved approaches are needed to deliver these drugs, particularly to cartilage tissue, which is avascular and contains a dense matrix of collagens and negatively charged aggrecan proteoglycans. Cationic delivery vehicles have been shown to effectively penetrate cartilage, but these studies have thus far largely focused on proteins or nanoparticles, and the effects of macromolecular architectures have not yet been explored. Described here is the synthesis of a small library of polycations composed of N-(2-hydroxypropyl)methacrylamide (HPMA) and N-(3-aminopropyl)methacrylamide (APMA) with linear, 4-arm, or 8-arm structures and varying degrees of polymerization (DP) by reversible addition fragmentation chain-transfer (RAFT) polymerization. Uptake and retention of the polycations in bovine articular cartilage was assessed. While all polycations penetrated cartilage, uptake and retention generally increased with DP before decreasing for the highest DP. In addition, uptake and retention were higher for the linear polycations compared to the 4-arm and 8-arm polycations. In general, the polycations were well tolerated by bovine chondrocytes, but the highest DP polycations imparted greater cytotoxicity. Overall, this study reveals that linear polymer architectures may be more favorable for binding to the cartilage matrix and that the DP can be tuned to maximize uptake while minimizing cytotoxicity.
Collapse
Affiliation(s)
- Jue Gong
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B7, Canada.
| | - Jordan Nhan
- Department of Chemical and Biological Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
| | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
| | - Elizabeth R Gillies
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B7, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B9, Canada
| |
Collapse
|
5
|
Le Z, Pan Q, He Z, Liu H, Shi Y, Liu L, Liu Z, Ping Y, Chen Y. Direct Cytosolic Delivery of Proteins and CRISPR-Cas9 Genome Editing by Gemini Amphiphiles via Non-Endocytic Translocation Pathways. ACS CENTRAL SCIENCE 2023; 9:1313-1326. [PMID: 37521791 PMCID: PMC10375873 DOI: 10.1021/acscentsci.3c00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 08/01/2023]
Abstract
Intracellular delivery of therapeutic biomacromolecules is often challenged by the poor transmembrane and limited endosomal escape. Here, we establish a combinatorial library composed of 150 molecular weight-defined gemini amphiphiles (GAs) to identify the vehicles that facilitate robust cytosolic delivery of proteins in vitro and in vivo. These GAs display similar skeletal structures but differential amphiphilicity by adjusting the length of alkyl tails, type of ionizable cationic heads, and hydrophobicity or hydrophilicity of a spacer. The top candidate is highly efficient in translocating a broad spectrum of proteins with various molecular weights and isoelectric points into the cytosol. Particularly, we notice that the entry mechanism is predominantly mediated via the lipid raft-dependent membrane fusion, bypassing the classical endocytic pathway that limits the cytosolic delivery efficiency of many presently available carriers. Remarkably, the top GA candidate is capable of delivering hard-to-deliver Cas9 ribonucleoprotein in vivo, disrupting KRAS mutation in the tumor-bearing mice to inhibit tumor growth and extend their survival. Our study reveals a GA-based small-molecule carrier platform for the direct cytosolic delivery of various types of proteins for therapeutic purposes.
Collapse
Affiliation(s)
- Zhicheng Le
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| | - Qi Pan
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zepeng He
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| | - Hong Liu
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| | - Yi Shi
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| | - Lixin Liu
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhijia Liu
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuan Ping
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongming Chen
- School
of Materials Science and Engineering, Key Laboratory for Polymeric
Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
6
|
Johnston BM, Grodzinsky AJ, Hammond PT. Charge shielding effects of PEG bound to NH 2-terminated PAMAM dendrimers - an experimental approach. SOFT MATTER 2023; 19:3033-3046. [PMID: 37038739 PMCID: PMC10131161 DOI: 10.1039/d2sm01698b] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cationic poly(amido amine) (PAMAM) dendrimers exhibit great potential for use in drug delivery, but their high charge density leads to an inherent cytotoxicity. To increase biocompatibility, many studies have attached poly(ethylene glycol) (PEG) chains to the dendrimer surface. It is unclear how these tethered PEG chains influence the physicochemical properties of the dendrimer. Here, we develop a fluorescence-based assay utilizing anionic biological tissue to quantify the electrostatic binding affinity of a library of PEG-PAMAM conjugates with various PEG chain lengths and grafting densities. We find that covalently bound PEG chains reduce the electrostatic binding affinity more significantly than what can be achieved through covalent bonds only. Contrary to previous thought, this reduction is not explained by the steric hindrance effects of PEG chains, suggesting that other, non-covalent interactions between PEG and PAMAM are present. Using acetylated PAMAM conjugates, we convert electrostatic binding affinity to the number of charged amines accessible to the physiological environment. These data, coupled with 1H-NMR, allows us to study more closely the non-covalent interactions between PEG and PAMAM. We find that increasing PEG chain length increases the number of non-covalent interactions. Additionally, at low grafting densities, increasing the number of PEG chains on the PAMAM surface also increases the non-covalent interactions. At higher grafting densities, however, PEG chains sterically repel one another, forcing chains to elongate away from the surface and reducing the number of interactions between PAMAM and individual PEG chains. The data presented here provides a framework for a more precise mechanistic understanding of how the length and density of tethered PEG chains on PAMAM dendrimers influence drug delivery properties.
Collapse
Affiliation(s)
- Brandon M Johnston
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139, USA.
- Koch Institute for Integrative Cancer Research, 500 Main St, Cambridge, MA, 02139, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Paula T Hammond
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, 02139, USA.
- Koch Institute for Integrative Cancer Research, 500 Main St, Cambridge, MA, 02139, USA
| |
Collapse
|
7
|
Haq-Siddiqi NA, Britton D, Kim Montclare J. Protein-engineered biomaterials for cartilage therapeutics and repair. Adv Drug Deliv Rev 2023; 192:114647. [PMID: 36509172 DOI: 10.1016/j.addr.2022.114647] [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: 06/05/2022] [Revised: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Cartilage degeneration and injury are major causes of pain and disability that effect millions, and yet treatment options for conditions like osteoarthritis (OA) continue to be mainly palliative or involve complete replacement of injured joints. Several biomaterial strategies have been explored to address cartilage repair either by the delivery of therapeutics or as support for tissue repair, however the complex structure of cartilage tissue, its mechanical needs, and lack of regenerative capacity have hindered this goal. Recent advances in synthetic biology have opened new possibilities for engineered proteins to address these unique needs. Engineered protein and peptide-based materials benefit from inherent biocompatibility and nearly unlimited tunability as they utilize the body's natural building blocks to fabricate a variety of supramolecular structures. The pathophysiology and needs of OA cartilage are presented here, along with an overview of the current state of the art and next steps for protein-engineered repair strategies for cartilage.
Collapse
Affiliation(s)
- Nada A Haq-Siddiqi
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Dustin Britton
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States; Department of Chemistry, New York University, New York 10003, United States; Department of Radiology, New York University Grossman School of Medicine, New York 10016, United States; Department of Biomaterials, NYU College of Dentistry, New York, NY 10010, United States; Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, United States.
| |
Collapse
|
8
|
Nagelli CV, Evans CH, De la Vega RE. Gene Delivery to Chondrocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:95-105. [PMID: 37052849 DOI: 10.1007/978-3-031-25588-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Delivering genes to chondrocytes offers new possibilities both clinically, for treating conditions that affect cartilage, and in the laboratory, for studying the biology of chondrocytes. Advances in gene therapy have created a number of different viral and non-viral vectors for this purpose. These vectors may be deployed in an ex vivo fashion, where chondrocytes are genetically modified outside the body, or by in vivo delivery where the vector is introduced directly into the body; in the case of articular and meniscal cartilage in vivo delivery is typically by intra-articular injection. Ex vivo delivery is favored in strategies for enhancing cartilage repair as these can be piggy-backed on existing cell-based technologies, such as autologous chondrocyte implantation, or used in conjunction with marrow-stimulating techniques such as microfracture. In vivo delivery to articular chondrocytes has proved more difficult, because the dense, anionic, extra-cellular matrix of cartilage limits access to the chondrocytes embedded within it. As Grodzinsky and colleagues have shown, the matrix imposes strict limits on the size and charge of particles able to diffuse through the entire depth of articular cartilage. Empirical observations suggest that the larger viral vectors, such as adenovirus (~100 nm), are unable to transduce chondrocytes in situ following intra-articular injection. However, adeno-associated virus (AAV; ~25 nm) is able to do so in horse joints. AAV is presently in clinical trials for arthritis gene therapy, and it will be interesting to see whether human chondrocytes are also transduced throughout the depth of cartilage by AAV following a single intra-articular injection. Viral vectors have been used to deliver genes to the intervertebral disk but there has been little research on gene transfer to chondrocytes in other cartilaginous tissues such as nasal, auricular or tracheal cartilage.
Collapse
|
9
|
Li J, Zhang H, Han Y, Hu Y, Geng Z, Su J. Targeted and responsive biomaterials in osteoarthritis. Theranostics 2023; 13:931-954. [PMID: 36793867 PMCID: PMC9925319 DOI: 10.7150/thno.78639] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/07/2022] [Indexed: 02/04/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease characterized by loss of articular cartilage and chronic inflammation, involving multiple cellular dysfunctions and tissue lesions. The non-vascular environment and dense cartilage matrix in the joints tend to block drug penetration, resulting in low drug bioavailability. There is a desire to develop safer and more effective OA therapies to meet the challenges of an aging world population in the future. Biomaterials have achieved satisfactory results in improving drug targeting, prolonging the duration of action, and achieving precision therapy. This article reviews the current basic understanding of the pathological mechanisms and clinical treatment dilemmas of OA, summarizes and discusses the advances for different kinds of targeted and responsive biomaterials in OA, seeking to provide new perspectives for the treatment of OA. Subsequently, limitations and challenges in clinical translation and biosafety are analyzed to guide the development of future therapeutic strategies for OA. As the need for precision medicine rises over time, emerging multifunctional biomaterials based on tissue targeting and controlled release will become an irreplaceable part of OA management.
Collapse
Affiliation(s)
- Jiadong Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Organoid Research Center, Shanghai University, Shanghai, 200444, China.,School of Medicine, Shanghai University, Shanghai 200444, China.,School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Yafei Han
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Yan Hu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.,Organoid Research Center, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
10
|
Warren MR, Bajpayee AG. Modeling Electrostatic Charge Shielding Induced by Cationic Drug Carriers in Articular Cartilage Using Donnan Osmotic Theory. Bioelectricity 2022; 4:248-258. [PMID: 36644714 PMCID: PMC9811830 DOI: 10.1089/bioe.2021.0026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background Positively charged drug carriers are rapidly emerging as a viable solution for long-standing challenges in delivery to dense, avascular, negatively charged tissues. These cationic carriers have demonstrated especially strong promise in targeting drugs to articular cartilage for osteoarthritis (OA) treatment. It is critical to evaluate the dose-dependent effects of their high intratissue uptake levels on charge-shielding of anionic matrix constituents, and the resulting changes in tissue osmotic swelling and mechanical integrity. Materials and Methods We use the ideal Donnan osmotic theory to derive a model for predicting intracartilage swelling pressures as a function of net charge (z) and equilibrium uptake of short-length, arginine-rich, multivalent, cationic peptide carriers (cationic peptide carriers [CPCs], z varied from +8 to +20) in cartilage samples with varying arthritic severities and fixed charge density (FCD). We use this model to determine the dose-dependent influence of CPCs on both physiological osmotic swelling pressures and compressive electrostatic moduli of cartilage in healthy and arthritic states. Results Under physiological conditions, the Donnan model predicted carrier-induced reductions in free swelling pressure between 8 and 29 kPa, and diminished compressive modulus by 20-68 kPa, both dependent on the net charge and uptake of CPCs. The magnitudes of deswelling and stiffness reduction increased monotonically with carrier uptake and net charge. Furthermore, predicted levels of deswelling by CPC charge shielding were amplified in tissues with reduced FCD (which model OA). Finally, the Donnan model predicted markedly higher reductions in tissue compressive modulus in hypotonic bathing salinity compared with physiological and hypertonic conditions. Conclusion This analysis demonstrates the importance of considering charge shielding as a likely adverse effect associated with uptake of cationic drug carriers into negatively charged tissues, especially in the case of damaged tissue. The simple modeling approach and principles described herein can inform the design of cationic drug delivery carriers and their clinical treatment regimens.
Collapse
Affiliation(s)
- Matthew R. Warren
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
- Department of Mechanical Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Xu XL, Xue Y, Ding JY, Zhu ZH, Wu XC, Song YJ, Cao YL, Tang LG, Ding DF, Xu JG. Nanodevices for deep cartilage penetration. Acta Biomater 2022; 154:23-48. [PMID: 36243371 DOI: 10.1016/j.actbio.2022.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease and is the main cause of chronic pain and functional disability in adults. Articular cartilage is a hydrated soft tissue that is composed of normally quiescent chondrocytes at a low density, a dense network of collagen fibrils with a pore size of 60-200 nm, and aggrecan proteoglycans with high-density negative charge. Although certain drugs, nucleic acids, and proteins have the potential to slow the progression of OA and restore the joints, these treatments have not been clinically applied owing to the lack of an effective delivery system capable of breaking through the cartilage barrier. Recently, the development of nanotechnology for delivery systems renders new ideas and treatment methods viable in overcoming the limited penetration. In this review, we focus on current research on such applications of nanotechnology, including exosomes, protein-based cationic nanocarriers, cationic liposomes/solid lipid nanoparticles, amino acid-based nanocarriers, polyamide derivatives-based nanocarriers, manganese dioxide, and carbon nanotubes. Exosomes are the smallest known nanoscale extracellular vesicles, and they can quickly deliver nucleic acids or proteins to the required depth. Through electrostatic interactions, nanocarriers with appropriate balance in cationic property and particle size have a strong ability to penetrate cartilage. Although substantial preclinical evidence has been obtained, further optimization is necessary for clinical transformation. STATEMENT OF SIGNIFICANCE: The dense cartilage matrix with high-negative charge was associated with reduced therapeutic effect in osteoarthritis patients with deep pathological changes. However, a systematic review in nanodevices for deep cartilage penetration is still lacking. Current approaches to assure penetration of nanosystems into the depth of cartilage were reviewed, including nanoscale extracellular vesicles from different cell lines and nanocarriers with appropriate balance in cationic property and size particle. Moreover, nanodevices entering clinical trials and further optimization were also discussed, providing important guiding significance to future research.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yan Xue
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Centre), School of Medicine, Tongji University, Shanghai 201613, China
| | - Jia-Ying Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhi-Heng Zhu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi-Chen Wu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yong-Jia Song
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue-Long Cao
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Long-Guang Tang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China.
| | - Dao-Fang Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Guang Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
12
|
Huang J, Liu Q, Xia J, Chen X, Xiong J, Yang L, Liang Y. Modification of mesenchymal stem cells for cartilage-targeted therapy. J Transl Med 2022; 20:515. [PMID: 36348497 PMCID: PMC9644530 DOI: 10.1186/s12967-022-03726-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/24/2022] [Indexed: 11/10/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by the destruction of the articular cartilage, sclerosis of the subchondral bone, and joint dysfunction. Its pathogenesis is attributed to direct damage and mechanical destruction of joint tissues. Mesenchymal stem cells (MSCs), suggested as a potential strategy for the treatment of OA, have shown therapeutic effects on OA. However, the specific fate of MSCs after intraarticular injection, including cell attachment, proliferation, differentiation, and death, is still unclear, and there is no guarantee that stem cells can be retained in the cartilage tissue to enact repair. Direct homing of MSCs is an important determinant of the efficacy of MSC-based cartilage repair. Recent studies have revealed that the unique homing capacity of MSCs and targeted modification can improve their ability to promote tissue regeneration. Here, we comprehensively review the homing effect of stem cells in joints and highlight progress toward the targeted modification of MSCs. In the future, developments of this targeting system that accelerate tissue regeneration will benefit targeted tissue repair.
Collapse
|
13
|
Oxymatrine Protects Chondrocytes against IL-1β-triggered Apoptosis in Vitro and Inhibits Osteoarthritis in Mice Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2745946. [PMID: 36204118 PMCID: PMC9532098 DOI: 10.1155/2022/2745946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022]
Abstract
Background Osteoarthritis (OA) is a multifactorial disease with various risk factors, resulting in the degeneration of articular cartilage and whole joints. However, to date, no effective disease-modifying therapy for OA has been developed. Oxymatrine (OMT) is associated with many pharmacological effects, including anti-inflammatory, antiapoptotic, and antioxidative properties. However, the role of OMT in OA remains unclear. Materials and Methods An IL-1β-induced chondrocyte model and anterior cruciate ligament transection (ACLT)-induced murine model of OA were constructed. The effect of OMT on chondrocyte viability was assessed using the CCK-8 assay. The protein level was assessed by Western blot analysis, and the apoptosis rate was assessed by flow cytometry in vitro and TUNEL staining in OA model mice. The effect of OMT on the degradation of articular cartilage in ACLT-induced OA mice was assessed by histological analysis. Results OMT at 0–2 mg/mL showed no conspicuous cytotoxicity on chondrocytes after 24 hours of incubation. OMT at 0.5, 1, and 2 mg/mL inhibited IL-1β-triggered apoptosis, upregulated MMP13, MMP9, and Col X, and upregulated Col II in chondrocytes in vitro. OMT represses the NF-κB signaling cascade in IL-1β-triggered chondrocytes in vitro. In an in vivo study, OMT decreased the apoptosis rate of chondrocytes and exerted a protective effect against the degradation of articular cartilage in ACLT-triggered OA mice. Conclusion OMT plays a protective role against chondrocyte injury induced by IL-1β in vitro or ACLT in vivo. OMT may play a role in chondrocytes during OA by inhibiting NF-κB signaling by decreasing the phosphorylation of p65 and IκB. OMT treatment may be a promising chondroprotective approach to delay OA cartilage progression.
Collapse
|
14
|
Warren MR, Vedadghavami A, Bhagavatula S, Bajpayee AG. Effects of polycationic drug carriers on the electromechanical and swelling properties of cartilage. Biophys J 2022; 121:3542-3561. [PMID: 35765244 PMCID: PMC9515003 DOI: 10.1016/j.bpj.2022.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/15/2022] Open
Abstract
Cationic nanocarriers offer a promising solution to challenges in delivering drugs to negatively charged connective tissues, such as to articular cartilage for the treatment of osteoarthritis (OA). However, little is known about the effects that cationic macromolecules may have on the mechanical properties of cartilage at high interstitial concentrations. We utilized arginine-rich cationic peptide carriers (CPCs) with varying net charge (from +8 to +20) to investigate the biophysical mechanisms of nanocarrier-induced alterations to cartilage biomechanical properties. We observed that CPCs increased the compressive modulus of healthy bovine cartilage explants by up to 70% and decreased the stiffness of glycosaminoglycan-depleted tissues (modeling OA) by 69%; in both cases, the magnitude of the change in stiffness correlated with the uptake of CPC charge variants. Next, we directly measured CPC-induced osmotic deswelling in cartilage tissue due to shielding of charge repulsions between anionic extracellular matrix constituents, with magnitudes of reductions between 36 and 64 kPa. We then demonstrated that electrostatic interactions were required for CPC-induced stiffening to occur, evidenced by no observed increase in tissue stiffness when measured in hypertonic bathing salinity. We applied a non-ideal Donnan osmotic model (under triphasic theory) to separate bulk modulus measurements into Donnan and non-Donnan components, which further demonstrated the conflicting charge-shielding and matrix-stiffening effects of CPCs. These results show that cationic drug carriers can alter tissue mechanical properties via multiple mechanisms, including the expected charge shielding as well as a novel stiffening phenomenon mediated by physical linkages. We introduce a model for how the magnitudes of these mechanical changes depend on tunable physical properties of the drug carrier, including net charge, size, and spatial charge distribution. We envision that the results and theory presented herein will inform the design of future cationic drug-delivery systems intended to treat diseases in a wide range of connective tissues.
Collapse
Affiliation(s)
- Matthew R Warren
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Sanjana Bhagavatula
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts; Department of Mechanical Engineering, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
15
|
Predicting Transport of Intra-Articularly Injected Growth Factor Fusion Proteins into Human Knee Joint Cartilage. Acta Biomater 2022; 153:243-259. [DOI: 10.1016/j.actbio.2022.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022]
|
16
|
Black RM, Flaman LL, Lindblom K, Chubinskaya S, Grodzinsky AJ, Önnerfjord P. Tissue catabolism and donor-specific dexamethasone response in a human osteochondral model of post-traumatic osteoarthritis. Arthritis Res Ther 2022; 24:137. [PMID: 35689293 PMCID: PMC9185927 DOI: 10.1186/s13075-022-02828-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/22/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Post-traumatic osteoarthritis (PTOA) does not currently have clinical prognostic biomarkers or disease-modifying drugs, though promising candidates such as dexamethasone (Dex) exist. Many challenges in studying and treating this disease stem from tissue interactions that complicate understanding of drug effects. We present an ex vivo human osteochondral model of PTOA to investigate disease effects on cartilage and bone homeostasis and discover biomarkers for disease progression and drug efficacy. METHODS Human osteochondral explants were harvested from normal (Collins grade 0-1) ankle talocrural joints of human donors (2 female, 5 male, ages 23-70). After pre-equilibration, osteochondral explants were treated with a single-impact mechanical injury and TNF-α, IL-6, and sIL-6R ± 100 nM Dex for 21 days and media collected every 2-3 days. Chondrocyte viability, tissue DNA content, and glycosaminoglycan (sGAG) percent loss to the media were assayed and compared to untreated controls using a linear mixed effects model. Mass spectrometry analysis was performed for both cartilage tissue and pooled culture medium, and the statistical significance of protein abundance changes was determined with the R package limma and empirical Bayes statistics. Partial least squares regression analyses of sGAG loss and Dex attenuation of sGAG loss against proteomic data were performed. RESULTS Injury and cytokine treatment caused an increase in the release of matrix components, proteases, pro-inflammatory factors, and intracellular proteins, while tissue lost intracellular metabolic proteins, which was mitigated with the addition of Dex. Dex maintained chondrocyte viability and reduced sGAG loss caused by injury and cytokine treatment by 2/3 overall, with donor-specific differences in the sGAG attenuation effect. Biomarkers of bone metabolism had mixed effects, and collagen II synthesis was suppressed with both disease and Dex treatment by 2- to 5-fold. Semitryptic peptides associated with increased sGAG loss were identified. Pro-inflammatory humoral proteins and apolipoproteins were associated with lower Dex responses. CONCLUSIONS Catabolic effects on cartilage tissue caused by injury and cytokine treatment were reduced with the addition of Dex in this osteochondral PTOA model. This study presents potential peptide biomarkers of early PTOA progression and Dex efficacy that can help identify and treat patients at risk of PTOA.
Collapse
Affiliation(s)
- Rebecca Mae Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Lisa L Flaman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Karin Lindblom
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Susan Chubinskaya
- Departments of Pediatrics, Orthopedic Surgery and Medicine (Section of Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | - Alan 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
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Dunshee LC, McDonough R, Price C, Kiick KL. Retention of peptide-based vesicles in murine knee joints after intra-articular injection. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
3D Printing for Cartilage Replacement: A Preliminary Study to Explore New Polymers. Polymers (Basel) 2022; 14:polym14051044. [PMID: 35267866 PMCID: PMC8914867 DOI: 10.3390/polym14051044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
The use of additive manufacturing technologies for biomedical applications must begin with the knowledge of the material to be used, by envisaging a very specific application rather than a more general aim. In this work, the preliminary study was focused on considering the cartilaginous tissue. This biological tissue exhibits different characteristics, such as thickness and mechanical properties, depending on its specific function in the body. Due to the lack of vascularization, cartilage is a supporting connective tissue with limited capacity for recovery and regeneration. For this reason, any approach, whether to repair/regenerate or as a total replacement, needs to fulfill the adequate mechanical and chemical properties of the surrounding native cartilage to be successful. This work aims to explore the possibility of using new polymers for cartilage total replacement approaches with polymeric materials processed with the specific 3D printing technique of fused filament fabrication (FFF). The materials studied were Nylon® 12 (PA12), already described for this purpose, and LAY-FOMM® 60 (FOMM). FOMM has not been described in the literature for biomedical purposes. Therefore, the chemical, thermal, swelling capacity, and mechanical properties of the filaments were thoroughly characterized to better understand the structure–properties–application relationships of this new polymer. In addition, as the FFF technology is temperature based, the properties were also evaluated in the printed specimens. Due to the envisaged application, the specimens were also characterized in the wet state. When comparing the obtained results with the properties of native cartilage, it was possible to conclude that: (i) PA12 exhibits low swelling capacity, while FOMM, in its dry and wet forms, has a higher swelling capacity, closer to that of native cartilage; (ii) the mechanical properties of the polymeric materials, especially PA12, are higher than those of native cartilage; and (iii) from the mechanical properties evaluated by ultra-micro hardness tests, the values for FOMM indicate that this material could be a good alternative for cartilage replacement in older patients. This preliminary study, essentially devoted to expanding the frontiers of the current state of the art of new polymeric materials, provides valuable indications for future work targeting the envisaged applications.
Collapse
|
19
|
Spatial configuration of charge and hydrophobicity tune particle transport through mucus. Biophys J 2022; 121:277-287. [PMID: 34951982 PMCID: PMC8790233 DOI: 10.1016/j.bpj.2021.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 10/26/2021] [Accepted: 12/16/2021] [Indexed: 01/21/2023] Open
Abstract
Mucus is a selectively permeable hydrogel that protects wet epithelia from pathogen invasion and poses a barrier to drug delivery. Determining the parameters of a particle that promote or prevent passage through mucus is critical, as it will enable predictions about the mucosal passage of pathogens and inform the design of therapeutics. The effect of particle net charge and size on mucosal transport has been characterized using simple model particles; however, predictions of mucosal passage remain challenging. Here, we utilize rationally designed peptides to examine the integrated contributions of charge, hydrophobicity, and spatial configuration on mucosal transport. We find that net charge does not entirely predict transport. Specifically, for cationic peptides, the inclusion of hydrophobic residues and the position of charged and hydrophobic residues within the peptide impact mucosal transport. We have developed a simple model of mucosal transport that predicts how previously unexplored amino acid sequences achieve slow versus fast passage through mucus. This model may be used as a basis to predict transport behavior of natural peptide-based particles, such as antimicrobial peptides or viruses, and assist in the engineering of synthetic sequences with desired transport properties.
Collapse
|
20
|
Day GJ, Zhang WH, Carter BM, Xiao W, Sambrook MR, Perriman AW. A Rationally Designed Supercharged Protein-Enzyme Chimera Self-Assembles In Situ to Yield Bifunctional Composite Textiles. ACS APPLIED MATERIALS & INTERFACES 2021; 13:60433-60445. [PMID: 34894651 DOI: 10.1021/acsami.1c18857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Catalytically active materials for the enhancement of personalized protective equipment (PPE) could be advantageous to help alleviate threats posed by neurotoxic organophosphorus compounds (OPs). Accordingly, a chimeric protein comprised of a supercharged green fluorescent protein (scGFP) and phosphotriesterase from Agrobacterium radiobacter (arPTE) was designed to drive the polymer surfactant (S-)-mediated self-assembly of microclusters to produce robust, enzymatically active materials. The chimera scGFP-arPTE was structurally characterized via circular dichroism spectroscopy and synchrotron radiation small-angle X-ray scattering, and its biophysical properties were determined. Significantly, the chimera exhibited greater thermal stability than the native constituent proteins, as well as a higher catalytic turnover number (kcat). Furthermore, scGFP-arPTE was electrostatically complexed with monomeric S-, driving self-assembly into [scGFP-arPTE][S-] nanoclusters, which could be dehydrated and cross-linked to yield enzymatically active [scGFP-arPTE][S-] porous films with a high-order structure. Moreover, these clusters could self-assemble within cotton fibers to generate active composite textiles without the need for the pretreatment of the fabrics. Significantly, the resulting materials maintained the biophysical activities of both constituent proteins and displayed recyclable and persistent activity against the nerve agent simulant paraoxon.
Collapse
Affiliation(s)
- Graham J Day
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - William H Zhang
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Ben M Carter
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Wenjin Xiao
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Mark R Sambrook
- CBR Division, Defence Science and Technology Laboratory (Dstl), Porton Down, Salisbury SP4 0JQ, United Kingdom
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
21
|
Intra-Articular Drug Delivery for Osteoarthritis Treatment. Pharmaceutics 2021; 13:pharmaceutics13122166. [PMID: 34959445 PMCID: PMC8703898 DOI: 10.3390/pharmaceutics13122166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease affecting millions of people worldwide. Currently, clinical nonsurgical treatments of OA are only limited to pain relief, anti-inflammation, and viscosupplementation. Developing disease-modifying OA drugs (DMOADs) is highly demanded for the efficient treatment of OA. As OA is a local disease, intra-articular (IA) injection directly delivers drugs to synovial joints, resulting in high-concentration drugs in the joint and reduced side effects, accompanied with traditional oral or topical administrations. However, the injected drugs are rapidly cleaved. By properly designing the drug delivery systems, prolonged retention time and targeting could be obtained. In this review, we summarize the drugs investigated for OA treatment and recent advances in the IA drug delivery systems, including micro- and nano-particles, liposomes, and hydrogels, hoping to provide some information for designing the IA injected formulations.
Collapse
|
22
|
Ebada HM, Nasra MM, Nassra RA, Solaiman AA, Abdallah OY. Cationic nanocarrier of rhein based on hydrophobic ion pairing approach as intra-articular targeted regenerative therapy for osteoarthritis. Colloids Surf B Biointerfaces 2021; 211:112285. [PMID: 34942464 DOI: 10.1016/j.colsurfb.2021.112285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 11/17/2022]
Abstract
Cartilage deterioration is the hallmark of osteoarthritis (OA). Rapid clearance of intra-articularly injected drugs and inherent cartilage barrier properties represent enormous challenges facing the effective local OA therapy. Rhein (RH), a dihydroxy-anthraquinone acid molecule, possess a potential chondroprotective effect. However, RH suffers from poor oral bioavailability besides its gastrointestinal side effects. Herein, for the first time, we exploited cationic carriers to target anionic cartilage matrix to create a RH-reservoir within the cartilage matrix, improving RH therapeutic efficacy with reduced side effects. Firstly, we improved RH lipophilic characteristics employing hydrophobic ion pairing (HIP) to be efficiently loaded within lipid nanoparticles with slow-release properties. RH-HIP integrated solid lipid nanoparticles (RH-SLNs) rapidly penetrated through cartilage tissue and lasted for 3 weeks into healthy and arthritic rat joints. Furthermore, RH-SLNs significantly inhibited inflammatory response, oxidative stress and cartilage deterioration in MIA-arthritic rats. In conclusion, intra-articular cationic RH-SLNs represented a meaningful step towards OA therapy.
Collapse
Affiliation(s)
- Heba Mk Ebada
- Central Lab, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt.
| | - Maha Ma Nasra
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Rasha A Nassra
- Department of Medical Biochemistery, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Amany A Solaiman
- Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
23
|
Horn JM, Obermeyer AC. Genetic and Covalent Protein Modification Strategies to Facilitate Intracellular Delivery. Biomacromolecules 2021; 22:4883-4904. [PMID: 34855385 PMCID: PMC9310055 DOI: 10.1021/acs.biomac.1c00745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein-based therapeutics represent a rapidly growing segment of approved disease treatments. Successful intracellular delivery of proteins is an important precondition for expanded in vivo and in vitro applications of protein therapeutics. Direct modification of proteins and peptides for improved cytosolic translocation are a promising method of increasing delivery efficiency and expanding the viability of intracellular protein therapeutics. In this Review, we present recent advances in both synthetic and genetic protein modifications for intracellular delivery. Active endocytosis-based and passive internalization pathways are discussed, followed by a review of modification methods for improved cytosolic delivery. After establishing how proteins can be modified, general strategies for facilitating intracellular delivery, such as chemical supercharging or inclusion of cell-penetrating motifs, are covered. We then outline protein modifications that promote endosomal escape. We finally examine the delivery of two potential classes of therapeutic proteins, antibodies and associated antibody fragments, and gene editing proteins, such as cas9.
Collapse
|
24
|
Black RM, Wang Y, Struglics A, Lorenzo P, Chubinskaya S, Grodzinsky AJ, Önnerfjord P. Proteomic clustering reveals the kinetics of disease biomarkers in bovine and human models of post-traumatic osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3. [DOI: 10.1016/j.ocarto.2021.100191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
25
|
An artificial membrane binding protein-polymer surfactant nanocomplex facilitates stem cell adhesion to the cartilage extracellular matrix. Biomaterials 2021; 276:120996. [PMID: 34280823 DOI: 10.1016/j.biomaterials.2021.120996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/12/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
One of the major challenges within the emerging field of injectable stem cell therapies for articular cartilage (AC) repair is the retention of sufficient viable cell numbers at the site of injury. Even when delivered via intra-articular injection, the number of stem cells retained at the target is often low and declines rapidly over time. To address this challenge, an artificial plasma membrane binding nanocomplex was rationally designed to provide human mesenchymal stem cells (hMSCs) with increased adhesion to articular cartilage tissue. The nanocomplex comprises the extracellular matrix (ECM) binding peptide of a placenta growth factor-2 (PlGF-2) fused to a supercharged green fluorescent protein (scGFP), which was electrostatically conjugated to anionic polymer surfactant chains to yield [S-]scGFP_PlGF2. The [S-]scGFP_PlGF2 nanocomplex spontaneously inserts into the plasma membrane of hMSCs, is not cytotoxic, and does not inhibit differentiation. The nanocomplex-modified hMSCs showed a significant increase in affinity for immobilised collagen II, a key ECM protein of cartilage, in both static and dynamic cell adhesion assays. Moreover, the cells adhered strongly to bovine ex vivo articular cartilage explants resulting in high cell numbers. These findings suggest that the re-engineering of hMSC membranes with [S-]scGFP_PlGF2 could improve the efficacy of injectable stem cell-based therapies for the treatment of damaged articular cartilage.
Collapse
|
26
|
Liang Y, Xu X, Xu L, Prasadam I, Duan L, Xiao Y, Xia J. Non-surgical osteoarthritis therapy, intra-articular drug delivery towards clinical applications. J Drug Target 2021; 29:609-616. [PMID: 33356642 DOI: 10.1080/1061186x.2020.1870231] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA)is a common orthopaedic disease in middle-aged and aged people. To date, no disease-modifying drug is available to prevent the progression of OA. Surgical treatment of OA has complications such as pain and high costs with increased risk of post-operative infections. An intra-articular drug delivery is a conservative treatment method to apply therapeutic composites directly into the OA joint cavity. This method has an advantage to improve the bioavailability of therapeutics and hence is a widely preferred choice to test novel disease-modifying drug targets for OA. Herein, we summarised and discussed the current status of intra-articular therapy for OA treatment as well as outlined drug delivery of small molecular, protein and gene delivery for OA therapy. Currently, new targeted nano-based drug delivery systems, including nanoparticles, exosomes and hydrogel formulations under investigation for OA treatment via intra-articular injection are also addressed. The emerging trend demonstrates that intra-articular drug delivery has vast prospects for the clinical selective treatment of OA. The rational application of intra-articular injection of drugs and biological agents will be of great significance for alleviating the patients with OA, improving their quality of life, delaying surgery, and reducing the disease burden of OA.
Collapse
Affiliation(s)
- Yujie Liang
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Chemistry, The Chinese University of Hong Kong, Shatin, China.,Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare & Shenzhen Institute of Mental Health, Shenzhen, China
| | - Xiao Xu
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Limei Xu
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Indira Prasadam
- Institute of Health and Biomedical Innovation, Faculty of Science and Engineering, Queensland University of Technology, Kelvin Grove Campus, Brisbane, Australia
| | - Li Duan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Faculty of Science and Engineering, Queensland University of Technology, Kelvin Grove Campus, Brisbane, Australia
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
27
|
Han Q, Ryan TM, Rosado CJ, Drummond CJ, Greaves TL. Effect of ionic liquids on the fluorescence properties and aggregation of superfolder green fluorescence protein. J Colloid Interface Sci 2021; 591:96-105. [PMID: 33596505 DOI: 10.1016/j.jcis.2021.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/24/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Proteins generally tend to aggregate with less desirable properties in numerous solvents, which is one of the major challenges in the development of solvents for functional proteins. This work aims to utilize fluorescence spectroscopy and small angle X-ray scattering (SAXS) to understand the effects of ionic liquids (ILs) on the fluorescence and aggregation behavior of superfolder green fluorescent protein (sfGFP). The studied ILs consisted of four different anions coupled with primary, tertiary and quaternary ammonium cations. The results show that the chromophore fluorescence was generally maintained in 1 mol% IL-water mixtures, then decreased with increasing IL concentration. We primarily employed the pseudo-radius of gyration (pseudo-Rg) to evaluate sfGFP aggregation. The sfGFP was less aggregated with nitrate-based ILs compared to in buffer, and more aggregated in the mesylate-based ILs. Further, we show that the polyol additives of glycerol and glucose in IL-water mixtures slightly decreased the sfGFP propensity to aggregate. Size-exclusion chromatography (SEC)-SAXS was used to characterize the monomeric sfGFP in ethylammonium nitrate (EAN) and triethylammonium mesylate (TEAMs)-water mixtures. The presence of 1 mol% TEAMs maintained the sfGFP fluorescence, promoted the compact structure, but slightly increased the amount of large aggregates, which contrasted with that of EAN.
Collapse
Affiliation(s)
- Qi Han
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, VIC 3000, Australia
| | - Timothy M Ryan
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Carlos J Rosado
- Department of Diabetes, Central Clinical School, Monash University, VIC 3004, Australia
| | - Calum J Drummond
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, VIC 3000, Australia
| | - Tamar L Greaves
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, VIC 3000, Australia.
| |
Collapse
|
28
|
Zhang X, Wu S, Zhu Y, Chu CQ. Exploiting Joint-Resident Stem Cells by Exogenous SOX9 for Cartilage Regeneration for Therapy of Osteoarthritis. Front Med (Lausanne) 2021; 8:622609. [PMID: 33681252 PMCID: PMC7928416 DOI: 10.3389/fmed.2021.622609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/20/2021] [Indexed: 12/27/2022] Open
Abstract
The lack of effective treatment options for osteoarthritis (OA) is mostly due to the very limited regenerative capacity of articular cartilage. Mesenchymal stem cells (MSCs) have been most extensively explored for cell-based therapy to induce cartilage regeneration for OA. However, current in vitro expanded MSC-based approaches have significant drawbacks. On the other hand, osteoarthritic joints contain chondrocyte progenitors and MSCs in several niches which have the potential yet fail to differentiate into chondrocytes for cartilage regeneration. One of the underlying mechanisms of the failure is that these chondrocyte progenitors and MSCs in OA joints are deficient in the activity of chondrogenic transcription factor SOX9 (SRY-type high-mobility group box-9). Thereby, replenishing with exogenous SOX9 would reactivate the potential of these stem cells to differentiate into chondrocytes. Cell-permeable, super-positively charged SOX9 (scSOX9) protein is able to promote hyaline-like cartilage regeneration by inducing chondrogenic differentiation of bone marrow derived MSCs in vivo. This scSOX9 protein can be administered into osteoarthritic joints by intra-articular injection. This one-step, cell-free supplement of exogenous SOX9 may harness the regenerative potential of the intrinsic MSCs within the joint cavity to stimulate cartilage regeneration in OA.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States.,Section of Rheumatology, VA Portland Health Care System, Portland, OR, United States
| | - Shili Wu
- Vivoscript, Inc., Irvine, CA, United States
| | - Yong Zhu
- Vivoscript, Inc., Irvine, CA, United States
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States.,Section of Rheumatology, VA Portland Health Care System, Portland, OR, United States
| |
Collapse
|
29
|
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: 4.3] [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
|
30
|
Abstract
PURPOSE OF REVIEW Osteoarthritis is associated with severe joint pain, inflammation, and cartilage degeneration. Drugs injected directly into intra-articular joint space clear out rapidly providing only short-term benefit. Their transport into cartilage to reach cellular targets is hindered by the tissue's dense, negatively charged extracellular matrix. This has limited, despite strong preclinical data, the clinical translation of osteoarthritis drugs. Recent work has focused on developing intra-joint and intra-cartilage targeting drug delivery systems (DDS) to enable long-term therapeutic response, which is presented here. RECENT FINDINGS Synovial joint targeting hybrid systems utilizing combinations of hydrogels, liposomes, and particle-based carriers are in consideration for pain-inflammation relief. Cartilage penetrating DDS target intra-cartilage constituents like aggrecans, collagen II, and chondrocytes such that drugs can reach their cellular and intra-cellular targets, which can enable clinical translation of disease-modifying osteoarthritis drugs including gene therapy. SUMMARY Recent years have witnessed significant increase in both fundamental and clinical studies evaluating DDS for osteoarthritis. Steroid encapsulating polymeric microparticles for longer lasting pain relief were recently approved for clinical use. Electrically charged biomaterials for intra-cartilage targeting have shown promising disease-modifying response in preclinical models. Clinical trials evaluating safety of viral vectors are ongoing whose success can pave the way for gene therapy as osteoarthritis treatment.
Collapse
Affiliation(s)
- Shikhar Mehta
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Tengfei He
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Mechanical & Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Intra-articular injection of anti-inflammatory peptide-loaded glycol chitosan/fucoidan nanogels to inhibit inflammation and attenuate osteoarthritis progression. Int J Biol Macromol 2020; 170:469-478. [PMID: 33359610 DOI: 10.1016/j.ijbiomac.2020.12.158] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/14/2020] [Accepted: 12/20/2020] [Indexed: 01/20/2023]
Abstract
Glycol chitosan/fucoidan nanogels loaded with anti-inflammatory peptide KAFAK (GC/Fu@KAFAK NGs) were fabricated based on the electrostatic interaction and genipin cross-linking methods. The prepared NGs had an average size of 286.3 ± 5.0 nm and positive surface charge of 14.0 ± 0.2 mV. The anti-inflammatory and chondro-protective effects of GC/Fu@KAFAK NGs were evaluated on interlecukin-1β (IL-1β)-stimulated rat chondrocytes. We found that GC/Fu@KAFAK NGs not only inhibited the expression of inflammatory factors interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), but also enhanced the expression of chondrogenic markers type II collagen, aggrecan, and Sox9. More importantly, in rat osteoarthritis (OA) model, the intra-articular (IA) injection of GC/Fu@KAFAK NGs reduced glycosaminoglycan loss and diminished inflammatory cytokine release. In addition, GC/Fu@KAFAK NGs showed good biocompatibility both in vitro and in vivo. In conclusion, IA inject-able GC/Fu@KAFAK NGs might have great potential in OA treatment.
Collapse
|
32
|
Ngo L, Knothe Tate ML. Osteoarthritis: New Strategies for Transport and Drug Delivery Across Length Scales. ACS Biomater Sci Eng 2020; 6:6009-6020. [PMID: 33449636 DOI: 10.1021/acsbiomaterials.0c01081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Osteoarthritis (OA) is the fourth leading cause of disability in adults. Yet, few viable pharmaceutical options exist for pain abatement and joint restoration, aside from joint replacement at late and irreversible stages of the disease. From the first onset of OA, as joint pain increases, individuals with arthritis increasingly reach for drug delivery solutions, from taking oral glycosaminoglycans (GAGs) bought over the counter from retail stores (e.g., Costco) to getting injections of viscous, GAG-containing synovial fluid supplement in the doctor's office. Little is known regarding the efficacy of delivery mode and/or treatment by such disease-modulating agents. This Review addresses the interplay of mechanics and biology on drug delivery to affected joints, which has profound implications for molecular transport in joint health and (patho)physiology. Multiscale systems biology approaches lend themselves to understand the relationship between the cell and joint health in OA and other joint (patho)physiologies. This Review first describes OA-related structural and functional changes in the context of the multilength scale anatomy of articular joints. It then summarizes and categorizes, by size and charge, published molecular transport studies, considering changes in permeability induced through inflammatory pathways. Finally, pharmacological interventions for OA are outlined in the context of molecular weights and modes of drug delivery. Taken together, the current state-of-the-art points to a need for new drug delivery strategies that harness systems-based interactions underpinning molecular transport and maintenance of joint structure and function at multiple length scales from molecular agents to cells, tissues, and tissue compartments which together make up articular joints. Cutting edge and cross-length and -time scale imaging represents a key discovery enabling technology in this process.
Collapse
Affiliation(s)
- Lucy Ngo
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Melissa L Knothe Tate
- Inaugural Paul Trainor Chair of Biomedical Engineering, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
33
|
Vedadghavami A, Zhang C, Bajpayee AG. Overcoming negatively charged tissue barriers: Drug delivery using cationic peptides and proteins. NANO TODAY 2020; 34:100898. [PMID: 32802145 PMCID: PMC7425807 DOI: 10.1016/j.nantod.2020.100898] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Negatively charged tissues are ubiquitous in the human body and are associated with a number of common diseases yet remain an outstanding challenge for targeted drug delivery. While the anionic proteoglycans are critical for tissue structure and function, they make tissue matrix dense, conferring a high negative fixed charge density (FCD) that makes drug penetration through the tissue deep zones and drug delivery to resident cells extremely challenging. The high negative FCD of these tissues is now being utilized by taking advantage of electrostatic interactions to create positively charged multi-stage delivery methods that can sequentially penetrate through the full thickness of tissues, create a drug depot and target cells. After decades of work on attempting delivery using strong binding interactions, significant advances have recently been made using weak and reversible electrostatic interactions, a characteristic now considered essential to drug penetration and retention in negatively charged tissues. Here we discuss these advances using examples of negatively charged tissues (cartilage, meniscus, tendons and ligaments, nucleus pulposus, vitreous of eye, mucin, skin), and delve into how each of their structures, tissue matrix compositions and high negative FCDs create barriers to drug entry and explore how charge interactions are being used to overcome these barriers. We review work on tissue targeting cationic peptide and protein-based drug delivery, compare and contrast drug delivery designs, and also present examples of technologies that are entering clinical trials. We also present strategies on further enhancing drug retention within diseased tissues of lower FCD by using synergistic effects of short-range binding interactions like hydrophobic and H-bonds that stabilize long-range charge interactions. As electrostatic interactions are incorporated into design of drug delivery materials and used as a strategy to create properties that are reversible, tunable and dynamic, bio-electroceuticals are becoming an exciting new direction of research and clinical work.
Collapse
Affiliation(s)
- Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Department of Mechanical Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
34
|
Black RM, Wang Y, Struglics A, Lorenzo P, Tillgren V, Rydén M, Grodzinsky AJ, Önnerfjord P. Proteomic analysis reveals dexamethasone rescues matrix breakdown but not anabolic dysregulation in a cartilage injury model. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2. [PMID: 34322675 DOI: 10.1016/j.ocarto.2020.100099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objectives In this exploratory study, we used discovery proteomics to follow the release of proteins from bovine knee articular cartilage in response to mechanical injury and cytokine treatment. We also studied the effect of the glucocorticoid Dexamethasone (Dex) on these responses. Design Bovine cartilage explants were treated with either cytokines alone (10 ng/ml TNFα, 20 ng/ml IL-6, 100 ng/ml sIL-6R), a single compressive mechanical injury, cytokines and injury, or no treatment, and cultured in serum-free DMEM supplemented with 1% ITS for 22 days. All samples were incubated with or without addition of 100 nM Dex. Mass spectrometry and western blot analyses were performed on medium samples for the identification and quantification of released proteins. Results We identified 500 unique proteins present in all three biological replicates. Many proteins involved in the catabolic response of cartilage degradation had increased release after inflammatory stress. Dex rescued many of these catabolic effects. The release of some proteins involved in anabolic and chondroprotective processes was inconsistent, indicating differential effects on processes that may protect cartilage from injury. Dex restored only a small fraction of these to the control state, while others had their effects exacerbated by Dex exposure. Conclusions We identified proteins that were released upon cytokine treatment which could be potential biomarkers of the inflammatory contribution to cartilage degradation. We also demonstrated the imperfect rescue of Dex on the effects of cartilage degradation, with many catabolic factors being reduced, while other anabolic or chondroprotective processes were not.
Collapse
Affiliation(s)
- Rebecca Mae Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - André Struglics
- Department of Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Pilar Lorenzo
- Department of Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Viveka Tillgren
- Department of Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Martin Rydén
- Department of Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Alan 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
| | - Patrik Önnerfjord
- Department of Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Magana JR, Sproncken CCM, Voets IK. On Complex Coacervate Core Micelles: Structure-Function Perspectives. Polymers (Basel) 2020; 12:E1953. [PMID: 32872312 PMCID: PMC7565781 DOI: 10.3390/polym12091953] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/31/2022] Open
Abstract
The co-assembly of ionic-neutral block copolymers with oppositely charged species produces nanometric colloidal complexes, known, among other names, as complex coacervates core micelles (C3Ms). C3Ms are of widespread interest in nanomedicine for controlled delivery and release, whilst research activity into other application areas, such as gelation, catalysis, nanoparticle synthesis, and sensing, is increasing. In this review, we discuss recent studies on the functional roles that C3Ms can fulfil in these and other fields, focusing on emerging structure-function relations and remaining knowledge gaps.
Collapse
Affiliation(s)
| | | | - Ilja K. Voets
- Laboratory of Self-Organizing Soft Matter, Department of Chemical Engineering and Chemistry and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands; (J.R.M.); (C.C.M.S.)
| |
Collapse
|
36
|
Komorek P, Wałek M, Jachimska B. Mechanism of lysozyme adsorption onto gold surface determined by quartz crystal microbalance and surface plasmon resonance. Bioelectrochemistry 2020; 135:107582. [PMID: 32535493 DOI: 10.1016/j.bioelechem.2020.107582] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 01/23/2023]
Abstract
In this study, the physicochemical characterization of lysozyme adsorbed on gold was investigated. Through the use of MP-SPR it was possible to establish that the orientation of molecules changes from side-on to between or end-on with increasing surface coverage. The data confirms that the process of adsorption is driven primarily by electrostatic interactions but also by hydrophobic forces. MP-SPR data was compared with the Random Sequential Adsorption model for a molecule with an ellipsoidal shape. Contact angle measurements showed that higher surface coverage also translates in more hydrophilic properties of obtained lysozyme layer. Comparison of CD and PM-IRRAS spectra in solution and adsorbed state respectively showed changes in the secondary structures of lysozyme. These changes are dependent on pH, but fundamentally they go in the direction of the increase of β-turn/random content with a simultaneous decrease in β-sheet fraction, which suggests that aggregation is not occurring. The combination of MP-SPR and QCM-D measurements allowed the estimation of the number of water molecules associated with the lysozymes films. It has been observed that hydration decreases from 70% in pH = 4 to 30% in pH = 11. This data indicates that hydration is driven mainly by the degree of protonation of lysozyme molecules.
Collapse
Affiliation(s)
- P Komorek
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Cracow, Poland
| | - M Wałek
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Cracow, Poland
| | - B Jachimska
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Cracow, Poland.
| |
Collapse
|
37
|
Young CC, Vedadghavami A, Bajpayee AG. Bioelectricity for Drug Delivery: The Promise of Cationic Therapeutics. Bioelectricity 2020; 2:68-81. [PMID: 32803148 DOI: 10.1089/bioe.2020.0012] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biological systems overwhelmingly comprise charged entities generating electrical activity that can have significant impact on biological structure and function. This intrinsic bio-electrical activity can also be harnessed for overcoming the tissue matrix and cell membrane barriers, which have been outstanding challenges for targeted drug delivery, by using rationally designed cationic carriers. The weak and reversible long-range electrostatic interactions with fixed negatively charged groups facilitate electro-diffusive transport of cationic therapeutics through full-tissue thickness to effectively reach intra-tissue, cellular, and intracellular target sites. This article presents a perspective on the promise of using rationally designed cationic biomaterials in targeted drug delivery, the underlying charge-based mechanisms, and bio-transport phenomena while addressing outstanding concerns around toxicity and methods to mitigate them. We also discuss electrically charged drugs that are currently being evaluated in clinical trials and identify areas of further development that have the potential to usher in new treatments.
Collapse
Affiliation(s)
- Cameron C Young
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA.,Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Adebayo OO, Holyoak DT, van der Meulen MCH. Mechanobiological Mechanisms of Load-Induced Osteoarthritis in the Mouse Knee. J Biomech Eng 2020; 141:2736041. [PMID: 31209459 DOI: 10.1115/1.4043970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects millions of people worldwide, yet its disease mechanism is not clearly understood. Animal models have been established to study disease progression by initiating OA through modified joint mechanics or altered biological activity within the joint. However, animal models often do not have the capability to directly relate the mechanical environment to joint damage. This review focuses on a novel in vivo approach based on controlled, cyclic tibial compression to induce OA in the mouse knee. First, we discuss the development of the load-induced OA model, its different loading configurations, and other techniques used by research laboratories around the world. Next, we review the lessons learned regarding the mechanobiological mechanisms of load-induced OA and relate these findings to the current understanding of the disease. Then, we discuss the role of specific genetic and cellular pathways involved in load-induced OA progression and the contribution of altered tissue properties to the joint response to mechanical loading. Finally, we propose using this approach to test the therapeutic efficacy of novel treatment strategies for OA. Ultimately, elucidating the mechanobiological mechanisms of load-induced OA will aid in developing targeted treatments for this disabling disease.
Collapse
Affiliation(s)
| | - Derek T Holyoak
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, 113 Weill Hall, Ithaca, NY 14853.,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853.,Research Division, Hospital for Special Surgery, New York, NY 10021 e-mail:
| |
Collapse
|
39
|
He T, Zhang C, Vedadghavami A, Mehta S, Clark HA, Porter RM, Bajpayee AG. Multi-arm Avidin nano-construct for intra-cartilage delivery of small molecule drugs. J Control Release 2019; 318:109-123. [PMID: 31843642 DOI: 10.1016/j.jconrel.2019.12.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 01/15/2023]
Abstract
Targeted drug delivery to joint tissues like cartilage remains a challenge that has prevented clinical translation of promising osteoarthritis (OA) drugs. Local intra-articular (IA) injections of drugs suffer from rapid clearance from the joint space and slow diffusive transport through the dense, avascular cartilage matrix comprised of negatively charged glycosaminoglycans (GAGs). Here we apply drug carriers that leverage electrostatic interactions with the tissue's high negative fixed charge density (FCD) for delivering small molecule drugs to cartilage cell and matrix sites. We demonstrate that a multi-arm cationic nano-construct of Avidin (mAv) with 28 sites for covalent drug conjugation can rapidly penetrate through the full thickness of cartilage in high concentration and have long intra-cartilage residence time in both healthy and arthritic cartilage via weak-reversible binding with negatively charged aggrecans. mAv's intra-cartilage mean uptake was found to be 112× and 33× the equilibration bath concentration in healthy and arthritic (50% GAG depleted) cartilage, respectively. mAv was conjugated with Dexamethasone (mAv-Dex), a broad-spectrum glucocorticoid, using a combination of hydrolysable ester linkers derived from succinic anhydride (SA), 3,3-dimethylglutaric anhydride (GA) and phthalic anhydride (PA) in 2:1:1 M ratio that enabled 50% drug release within 38.5 h followed by sustained release in therapeutic doses over 2 weeks. A single 10 μM low dose of controlled release mAv-Dex (2:1:1) effectively suppressed IL-1α-induced GAG loss, cell death and inflammatory response significantly better than unmodified Dex over 2 weeks in cartilage explant culture models of OA. With this multi-arm design, <1 μM Avidin was needed - a concentration which has been shown to be safe, preventing further GAG loss and cytotoxicity. A charge-based cartilage homing drug delivery platform like this can elicit disease modifying effects as well as facilitate long-term symptomatic pain and inflammation relief by enhancing tissue specificity and prolonging intra-cartilage residence time of OA drugs. This nano-construct thus has high translational potential for enabling intra-cartilage delivery of a broad array of small molecule OA drugs and their combinations to chondrocytes, enabling OA treatment with a single injection of low drug doses and eliminating toxicity issues associated with multiple high dose injections.
Collapse
Affiliation(s)
- Tengfei He
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Chenzhen Zhang
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Armin Vedadghavami
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Shikhar Mehta
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Heather A Clark
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA; Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA.
| | - Ryan M Porter
- Departments of Internal Medicine and Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Ambika G Bajpayee
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA; Mechanical Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
40
|
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
|
41
|
Evans CH. Catering to chondrocytes. Sci Transl Med 2019; 10:10/469/eaav7043. [PMID: 30487250 DOI: 10.1126/scitranslmed.aav7043] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
Abstract
An innovative strategy for delivering drugs to chondrocytes in situ offers new avenues for treating osteoarthritis (Geiger et al, this issue).
Collapse
Affiliation(s)
- Christopher H Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
42
|
Xiao W, Green TIP, Liang X, Delint RC, Perry G, Roberts MS, Le Vay K, Back CR, Ascione R, Wang H, Race PR, Perriman AW. Designer artificial membrane binding proteins to direct stem cells to the myocardium. Chem Sci 2019; 10:7610-7618. [PMID: 31588312 PMCID: PMC6764276 DOI: 10.1039/c9sc02650a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
We present a new cell membrane modification methodology where the inherent heart tissue homing properties of the infectious bacteria Streptococcus gordonii are transferred to human stem cells. This is achieved via the rational design of a chimeric protein-polymer surfactant cell membrane binding construct, comprising the cardiac fibronectin (Fn) binding domain of the bacterial adhesin protein CshA fused to a supercharged protein. Significantly, the protein-polymer surfactant hybrid spontaneously inserts into the plasma membrane of stem cells without cytotoxicity, instilling the cells with a high affinity for immobilized fibronectin. Moreover, we show that this cell membrane reengineering approach significantly improves retention and homing of stem cells delivered either intracardially or intravenously to the myocardium in a mouse model.
Collapse
Affiliation(s)
- Wenjin Xiao
- School of Cellular and Molecular Medicine , University of Bristol , BS8 1TD , UK .
| | - Thomas I P Green
- School of Cellular and Molecular Medicine , University of Bristol , BS8 1TD , UK .
- Bristol Centre for Functional Nanomaterials , University of Bristol , BS8 1FD , UK
| | - Xiaowen Liang
- The University of Queensland Diamantina Institute , The University of Queensland , Translational Research Institute , Woolloongabba , QLD 4102 , Australia
| | - Rosalia Cuahtecontzi Delint
- School of Cellular and Molecular Medicine , University of Bristol , BS8 1TD , UK .
- Bristol Centre for Functional Nanomaterials , University of Bristol , BS8 1FD , UK
| | - Guillaume Perry
- Sorbonne Université , Laboratoire d'Electronique et d'Electromagnétisme, L2E , F-75005 , Paris , France
| | - Michael S Roberts
- The University of Queensland Diamantina Institute , The University of Queensland , Translational Research Institute , Woolloongabba , QLD 4102 , Australia
- School of Pharmacy and Medical Science , University of South Australia , Adelaide , SA 5001 , Australia
| | - Kristian Le Vay
- School of Cellular and Molecular Medicine , University of Bristol , BS8 1TD , UK .
- Bristol Centre for Functional Nanomaterials , University of Bristol , BS8 1FD , UK
| | - Catherine R Back
- School of Biochemistry , University of Bristol , BS8 1TD , UK
- BrisSynBio Synthetic Biology Research Centre , University of Bristol , BS8 1TQ , UK
| | - Raimomdo Ascione
- Translational Biomedical Research Centre and Bristol Heart Institute , University of Bristol , Bristol , UK
| | - Haolu Wang
- The University of Queensland Diamantina Institute , The University of Queensland , Translational Research Institute , Woolloongabba , QLD 4102 , Australia
| | - Paul R Race
- School of Biochemistry , University of Bristol , BS8 1TD , UK
- BrisSynBio Synthetic Biology Research Centre , University of Bristol , BS8 1TQ , UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine , University of Bristol , BS8 1TD , UK .
- BrisSynBio Synthetic Biology Research Centre , University of Bristol , BS8 1TQ , UK
| |
Collapse
|
43
|
Vedadghavami A, Wagner EK, Mehta S, He T, Zhang C, Bajpayee AG. Cartilage penetrating cationic peptide carriers for applications in drug delivery to avascular negatively charged tissues. Acta Biomater 2019; 93:258-269. [PMID: 30529083 DOI: 10.1016/j.actbio.2018.12.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/19/2018] [Accepted: 12/04/2018] [Indexed: 02/01/2023]
Abstract
Drug delivery to avascular, negatively charged tissues like cartilage remains a challenge. The constant turnover of synovial fluid results in short residence time of administered drugs in the joint space and the dense negatively charged matrix of cartilage hinders their diffusive transport. Drugs are, therefore, unable to reach their cell and matrix targets in sufficient doses, and fail to elicit relevant biological response, which has led to unsuccessful clinical trials. The high negative fixed charge density (FCD) of cartilage, however, can be used to convert cartilage from a barrier to drug entry into a depot by making drugs positively charged. Here we design cartilage penetrating and binding cationic peptide carriers (CPCs) with varying net charge, spatial distribution and hydrophobicity to deliver large-sized therapeutics and investigate their electro-diffusive transport in healthy and arthritic cartilage. We showed that CPC uptake increased with increasing net charge up to +14 but dropped as charge increased further due to stronger binding interactions that hindered CPC penetrability and uptake showing that weak-reversible binding is key to enable their penetration through full tissue thickness. Even after 90% GAG depletion, while CPC +14 uptake reduced by over 50% but still had a significantly high value of 148× showing that intra-tissue long-range charge-based binding is further stabilized by short-range H-bond and hydrophobic interactions. The work presents an approach for rational design of cationic carriers based on tissue FCD and properties of macromolecules to be delivered. These design rules can be extended to drug delivery for other avascular, negatively charged tissues. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) remains an untreatable disease partly due to short joint residence time of drugs and a lack of delivery methods that can effectively target the dense, avascular, highly negatively charged cartilage tissue. In this study, we designed cartilage penetrating and binding cationic peptide carriers (CPCs) that, due to their optimal charge provide adequate electrical driving force to rapidly transport OA drugs into cartilage and reach their cell and matrix targets in therapeutic doses before drugs exit the joint space. This way cartilage is converted from being a barrier to drug entry into a drug depot that can provide sustained drug release for several weeks. This study also investigates synergistic effects of short-range H-bond and hydrophobic interactions in combination with long-range electrostatic interactions on intra-cartilage solute transport. The work provides rules for rational design of cartilage penetrating charge-based carriers depending on the net charge of tissue (normal versus arthritic), macromolecule to be delivered and whether the application is in drug delivery or tissue imaging.
Collapse
|
44
|
Horn JM, Kapelner RA, Obermeyer AC. Macro- and Microphase Separated Protein-Polyelectrolyte Complexes: Design Parameters and Current Progress. Polymers (Basel) 2019; 11:E578. [PMID: 30960562 PMCID: PMC6523202 DOI: 10.3390/polym11040578] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/18/2019] [Accepted: 03/23/2019] [Indexed: 01/02/2023] Open
Abstract
Protein-containing polyelectrolyte complexes (PECs) are a diverse class of materials, composed of two or more oppositely charged polyelectrolytes that condense and phase separate near overall charge neutrality. Such phase-separation can take on a variety of morphologies from macrophase separated liquid condensates, to solid precipitates, to monodispersed spherical micelles. In this review, we present an overview of recent advances in protein-containing PECs, with an overall goal of defining relevant design parameters for macro- and microphase separated PECs. For both classes of PECs, the influence of protein characteristics, such as surface charge and patchiness, co-polyelectrolyte characteristics, such as charge density and structure, and overall solution characteristics, such as salt concentration and pH, are considered. After overall design features are established, potential applications in food processing, biosensing, drug delivery, and protein purification are discussed and recent characterization techniques for protein-containing PECs are highlighted.
Collapse
Affiliation(s)
- Justin M Horn
- Department of Chemical Engineering, Columbia University, New York, NY 10027, USA.
| | - Rachel A Kapelner
- Department of Chemical Engineering, Columbia University, New York, NY 10027, USA.
| | - Allie C Obermeyer
- Department of Chemical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|