1
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
2
|
Zhang C, Pathrikar TV, Baby HM, Li J, Zhang H, Selvadoss A, Ovchinnikova A, Ionescu A, Chubinskaya S, Miller RE, Bajpayee AG. Charge-Reversed Exosomes for Targeted Gene Delivery to Cartilage for Osteoarthritis Treatment. SMALL METHODS 2024; 8:e2301443. [PMID: 38607953 PMCID: PMC11470115 DOI: 10.1002/smtd.202301443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/18/2024] [Indexed: 04/14/2024]
Abstract
Gene therapy has the potential to facilitate targeted expression of therapeutic proteins to promote cartilage regeneration in osteoarthritis (OA). The dense, avascular, aggrecan-glycosaminoglycan (GAG) rich negatively charged cartilage, however, hinders their transport to reach chondrocytes in effective doses. While viral vector mediated gene delivery has shown promise, concerns over immunogenicity and tumorigenic side-effects persist. To address these issues, this study develops surface-modified cartilage-targeting exosomes as non-viral carriers for gene therapy. Charge-reversed cationic exosomes are engineered for mRNA delivery by anchoring cartilage targeting optimally charged arginine-rich cationic motifs into the anionic exosome bilayer by using buffer pH as a charge-reversal switch. Cationic exosomes penetrated through the full-thickness of early-stage arthritic human cartilage owing to weak-reversible ionic binding with GAGs and efficiently delivered the encapsulated eGFP mRNA to chondrocytes residing in tissue deep layers, while unmodified anionic exosomes do not. When intra-articularly injected into destabilized medial meniscus mice knees with early-stage OA, mRNA loaded charge-reversed exosomes overcame joint clearance and rapidly penetrated into cartilage, creating an intra-tissue depot and efficiently expressing eGFP; native exosomes remained unsuccessful. Cationic exosomes thus hold strong translational potential as a platform technology for cartilage-targeted non-viral delivery of any relevant mRNA targets for OA treatment.
Collapse
Affiliation(s)
- Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Tanvi V. Pathrikar
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Helna M. Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Jun Li
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | | | - Andreia Ionescu
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical College, Chicago, IL 60612, USA
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
3
|
Millán Cotto HA, Pathrikar TV, Hakim B, Baby HM, Zhang H, Zhao P, Ansaripour R, Amini R, Carrier RL, Bajpayee AG. Cationic-motif-modified exosomes for mRNA delivery to retinal photoreceptors. J Mater Chem B 2024; 12:7384-7400. [PMID: 38946491 PMCID: PMC11323772 DOI: 10.1039/d4tb00849a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Topical treatment of vitreoretinal diseases remains a challenge due to slow corneal uptake and systemic clearance. Exosomes are emerging nanocarriers for drug delivery due to biocompatibility and cellular targeting properties. To apply them for retinal targeting via the topical route, exosomes must traverse various ocular barriers including the cornea, lens, vitreous humor (VH), and the retina itself. Here we engineered high-purity milk-derived exosomes by anchoring arginine-rich cationic motifs via PEG2000 lipid insertion on their surface. Modification enabled exosomes to use weak-reversible electrostatic interactions with anionic glycosaminoglycan (GAG) and water content of the tissue to enhance their transport rate and retention. Addition of cationic motifs neutralized the anionic surface charge of exosomes (-24 to -2 mV) without impacting size or morphology. Cationic-motif-modified exosomes exhibited two-fold faster steady state diffusivity through bovine corneas compared to unmodified exosomes. Fluorescence recovery after photobleaching confirmed that cationic-motif-modified exosomes can diffuse through VH without steric hindrance. In healthy VH, cationic-motif-modified exosomes demonstrated stronger binding resulting in three-fold lower average diffusivity that enhanced by six-fold in 50% GAG-depleted VH recapitulating advanced liquefaction. Cationic-motif-modified exosomes penetrated through the full-thickness of porcine retinal explants resulting in ten-fold higher uptake in photoreceptors and three-fold greater transfection with encapsulated eGFP mRNA compared to unmodified exosomes. Cationic-motif-modified exosomes are safe to use as they did not adversely affect the mechanical swelling properties of the cornea or lens nor impact retinal cell viability. Cationic-motif-modified exosomes, therefore, offer themselves as a cell-free nanocarrier platform for gene delivery to retinal photoreceptors potentially via the topical route.
Collapse
Affiliation(s)
| | | | - Bill Hakim
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Peng Zhao
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Ronak Ansaripour
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Rouzbeh Amini
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
4
|
Wu J, Roesger S, Jones N, Hu CMJ, Li SD. Cell-penetrating peptides for transmucosal delivery of proteins. J Control Release 2024; 366:864-878. [PMID: 38272399 DOI: 10.1016/j.jconrel.2024.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Enabling non-invasive delivery of proteins across the mucosal barriers promises improved patient compliance and therapeutic efficacies. Cell-penetrating peptides (CPPs) are emerging as a promising and versatile tool to enhance protein and peptide permeation across various mucosal barriers. This review examines the structural and physicochemical attributes of the nasal, buccal, sublingual, and oral mucosa that hamper macromolecular delivery. Recent development of CPPs for overcoming those mucosal barriers for protein delivery is summarized and analyzed. Perspectives regarding current challenges and future research directions towards improving non-invasive transmucosal delivery of macromolecules for ultimate clinical translation are discussed.
Collapse
Affiliation(s)
- Jiamin Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Sophie Roesger
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Natalie Jones
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Che-Ming J Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|
5
|
Zhang C, Zhang H, Millán Cotto HA, Boyer TL, Warren MR, Wang CM, Luchan J, Dhal PK, Carrier RL, Bajpayee AG. Milk exosomes anchored with hydrophilic and zwitterionic motifs enhance mucus permeability for applications in oral gene delivery. Biomater Sci 2024; 12:634-649. [PMID: 38047368 PMCID: PMC10842862 DOI: 10.1039/d3bm01089a] [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: 12/05/2023]
Abstract
Exosomes have emerged as a promising tool for the delivery of drugs and genetic materials, owing to their biocompatibility and non-immunogenic nature. However, challenges persist in achieving successful oral delivery due to their susceptibility to degradation in the harsh gastrointestinal (GI) environment and impeded transport across the mucus-epithelium barrier. To overcome these challenges, we have developed high-purity bovine milk exosomes (mExo) as a scalable and efficient oral drug delivery system, which can be customized by incorporating hydrophilic and zwitterionic motifs on their surface. In our study, we observed significantly improved transport rates by 2.5-4.5-fold in native porcine intestinal mucus after the introduction of hydrophilic and zwitterionic surface modifications, as demonstrated by transwell setup and fluorescence recovery after photobleaching (FRAP) analysis. Remarkably, mExo functionalized by a block peptide (BP), consisting of cationic and anionic amino acids arranged in blocks at the two ends, demonstrated superior tolerability in the acidic gastric environment (with a protein recovery rate of 84.8 ± 7.7%) and exhibited a 2.5-fold increase in uptake by intestinal epithelial cells. Furthermore, both mExo and mExo-BP demonstrated successful intracellular delivery of functional siRNA, resulting in up to 65% suppression of the target green fluorescence protein (GFP) gene expression at a low dose of siRNA (5 pmol) without causing significant toxicity. These findings highlight the immense potential of modifying mExo with hydrophilic and zwitterionic motifs for effective oral delivery of siRNA therapies.
Collapse
Affiliation(s)
- Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | | | - Timothy L Boyer
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Matthew R Warren
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Joshua Luchan
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | | | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
Chen X, Xu Z, Li T, Thakur A, Wen Y, Zhang K, Liu Y, Liang Q, Liu W, Qin JJ, Yan Y. Nanomaterial-encapsulated STING agonists for immune modulation in cancer therapy. Biomark Res 2024; 12:2. [PMID: 38185685 PMCID: PMC10773049 DOI: 10.1186/s40364-023-00551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 01/09/2024] Open
Abstract
The cGAS-STING signaling pathway has emerged as a critical mediator of innate immune responses, playing a crucial role in improving antitumor immunity through immune effector responses. Targeting the cGAS-STING pathway holds promise for overcoming immunosuppressive tumor microenvironments (TME) and promoting effective tumor elimination. However, systemic administration of current STING agonists faces challenges related to low bioavailability and potential adverse effects, thus limiting their clinical applicability. Recently, nanotechnology-based strategies have been developed to modulate TMEs for robust immunotherapeutic responses. The encapsulation and delivery of STING agonists within nanoparticles (STING-NPs) present an attractive avenue for antitumor immunotherapy. This review explores a range of nanoparticles designed to encapsulate STING agonists, highlighting their benefits, including favorable biocompatibility, improved tumor penetration, and efficient intracellular delivery of STING agonists. The review also summarizes the immunomodulatory impacts of STING-NPs on the TME, including enhanced secretion of pro-inflammatory cytokines and chemokines, dendritic cell activation, cytotoxic T cell priming, macrophage re-education, and vasculature normalization. Furthermore, the review offers insights into co-delivered nanoplatforms involving STING agonists alongside antitumor agents such as chemotherapeutic compounds, immune checkpoint inhibitors, antigen peptides, and other immune adjuvants. These platforms demonstrate remarkable versatility in inducing immunogenic responses within the TME, ultimately amplifying the potential for antitumor immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Tongfei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, 60637, Chicago, IL, USA
| | - Yu Wen
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Furong Laboratory, Central South University, 410008, Changsha, Hunan, China
| | - Kui Zhang
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, 60637, Chicago, IL, USA
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, 310022, Hangzhou, Zhejiang, China.
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
7
|
Arroyo-Urea EM, Muñoz-Hernando M, Leo-Barriga M, Herranz F, González-Paredes A. A quality by design approach for the synthesis of palmitoyl-L-carnitine-loaded nanoemulsions as drug delivery systems. Drug Deliv 2023; 30:2179128. [PMID: 36803136 PMCID: PMC10184586 DOI: 10.1080/10717544.2023.2179128] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Nanoemulsions (NE) are lipid nanocarriers that can efficiently load hydrophobic active compounds, like palmitoyl-L-carnitine (pC), used here as model molecule. The use of design of experiments (DoE) approach is a useful tool to develop NEs with optimized properties, requiring less experiments compared to trial-and-error approach. In this work, NE were prepared by the solvent injection technique and DoE using a two-level fractional factorial design (FFD) as model was implemented for designing pC-loaded NE. NEs were fully characterized by a combination of techniques, studying its stability, scalability, pC entrapment and loading capacity and biodistribution, which was studied ex-vivo after injection of fluorescent NEs in mice. We selected the optimal composition for NE, named pC-NEU, after analysis of four variables using DoE. pC-NEU incorporated pC in a very efficient manner, with high entrapment efficiency (EE) and loading capacity. pC-NEU did not change its initial colloidal properties stored at 4 °C in water during 120 days, nor in buffers with different pH values (5.3 and 7.4) during 30 days. Moreover, the scalability process did not affect NE properties and stability profile. Finally, biodistribution study showed that pC-NEU formulation was predominantly concentrated in the liver, with minimal accumulation in spleen, stomach, and kidneys.
Collapse
Affiliation(s)
- E M Arroyo-Urea
- Nanomedicine and Molecular Imaging group, Instituto de Química Médica-CSIC, Madrid, Spain
| | - María Muñoz-Hernando
- Nanomedicine and Molecular Imaging group, Instituto de Química Médica-CSIC, Madrid, Spain
| | - Marta Leo-Barriga
- Nanomedicine and Molecular Imaging group, Instituto de Química Médica-CSIC, Madrid, Spain
| | - Fernando Herranz
- Nanomedicine and Molecular Imaging group, Instituto de Química Médica-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Conexión Nanomedicina-CSIC, Madrid, Spain
| | - Ana González-Paredes
- Nanomedicine and Molecular Imaging group, Instituto de Química Médica-CSIC, Madrid, Spain.,Conexión Nanomedicina-CSIC, Madrid, Spain
| |
Collapse
|
8
|
Mehta S, Boyer TL, Akhtar S, He T, Zhang C, Vedadghavami A, Bajpayee AG. Sustained intra-cartilage delivery of interleukin-1 receptor antagonist using cationic peptide and protein-based carriers. Osteoarthritis Cartilage 2023; 31:780-792. [PMID: 36739939 PMCID: PMC10392024 DOI: 10.1016/j.joca.2023.01.573] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/20/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Blocking the interleukin-1 (IL-1) catabolic cascade following joint trauma can be achieved using its receptor antagonist, IL-1Ra. However, its clinical translation for osteoarthritis therapy has been unsuccessful due to its rapid joint clearance and lack of targeting and penetration into deep cartilage layers at therapeutic concentrations. Here, we target the high negative charge of cartilage aggrecan-glycosaminoglycans (GAGs) by attaching cationic carriers to IL-1Ra. IL-1Ra was conjugated to the cartilage targeting glycoprotein, Avidin, and a short length optimally charged cationic peptide carrier (CPC+14). It is hypothesized that electro-diffusive transport and binding properties of IL-1Ra-Avidin and IL-1Ra-CPC+14 will create intra-cartilage depots of IL-1Ra, resulting in long-term suppression of IL-1 catabolism with only a single administration. DESIGN IL-1Ra was conjugated to Avidin or CPC+14 using site specific maleimide linkers, and confirmed using gel electrophoresis, high-performance liquid chromatography (HPLC), and mass spectrometry. Intra-cartilage transport and retention of conjugates was compared with native IL-1Ra. Attenuation of IL-1 catabolic signaling with one-time dose of IL-1Ra-CPC+14 and IL-1Ra-Avidin was assessed over 16 days using IL-1α challenged bovine cartilage and compared with unmodified IL-1Ra. RESULTS Positively charged IL-1Ra penetrated through the full-thickness of cartilage, creating a drug depot. A single dose of unmodified IL-1Ra was not sufficient to attenuate IL-1-induced cartilage deterioration over 16 days. However, when delivered using Avidin, and to a greater extent CPC+14, IL-1Ra significantly suppressed cytokine induced GAG loss and nitrite release while improving cell metabolism and viability. CONCLUSION Charge-based cartilage targeting drug delivery systems hold promise as they can enable long-term therapeutic benefit with only a single dose.
Collapse
Affiliation(s)
- S Mehta
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - T L Boyer
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - S Akhtar
- Department of Biochemistry, Northeastern University, Boston, MA, USA.
| | - T He
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - C Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - A Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - A G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Mechanical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
9
|
Campbell J, Taghavi A, Preis A, Martin S, Skirtach AG, Franke J, Volodkin D, Vikulina A. Spontaneous shrinkage drives macromolecule encapsulation into layer-by-layer assembled biopolymer microgels. J Colloid Interface Sci 2023; 635:12-22. [PMID: 36577351 DOI: 10.1016/j.jcis.2022.12.115] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
HYPOTHESIS Recently, the anomalous shrinkage of surface-supported hyaluronate/poly-l-lysine (HA/PLL) microgels (µ-gels), which exceeds that reported for any other multilayer-based systems, has been reported [1]. The current study investigates the capability of these unique µ-gels for the encapsulation and retention of macromolecules, and proposes the shrinkage-driven assembly of biopolymer-based µ-gels as a novel tool for one-step surface biofunctionalization. EXPERIMENTS A set of dextrans (DEX) and their charged derivatives - carboxymethyl (CM)-DEX and diethylaminoethyl (DEAE)-DEX - has been utilized to evaluate the effects of macromolecular mass and net charge on µ-gel shrinkage and macromolecule entrapment. µ-gels formation on the surface of silicone catheters exemplifies their potential to tailor biointerfaces. FINDINGS Shrinkage-driven µ-gel formation strongly depends on the net charge and mass content of encapsulated macromolecules. Inclusion of neutral DEX decreases the degree of shrinkage several times, whilst charged DEXs adopt to the backbone of oppositely charged polyelectrolytes, resulting in shrinkage comparable to that of non-loaded µ-gels. Retention of CM-DEX in µ-gels is significantly higher compared to DEAE-DEX. These insights into the mechanisms of macromolecular entrapment into biopolymer-based µ-gels promotes fundamental understanding of molecular dynamics within the multilayer assemblies. Organization of biodegradable µ-gels at biomaterial surfaces opens avenues for their further exploitation in a diverse array of bioapplications.
Collapse
Affiliation(s)
- Jack Campbell
- Department of Chemistry and Forensics, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom; Bavarian Polymer Institute, Friedrich-Alexander-Universität Erlangen-Nürnberg, Dr.-Mack-Straße 77, 90762 Fürth, Germany
| | - Aaron Taghavi
- Department of Chemistry and Forensics, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Alexander Preis
- Institute for Factory Automation and Production Systems (FAPS), Friedrich-Alexander-Universität Erlangen-Nürnberg, Egerlandstr. 7-9, 91058 Erlangen, Germany
| | - Sina Martin
- Institute for Factory Automation and Production Systems (FAPS), Friedrich-Alexander-Universität Erlangen-Nürnberg, Egerlandstr. 7-9, 91058 Erlangen, Germany
| | - Andre G Skirtach
- Nano-Biotechnology Laboratory, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Jörg Franke
- Institute for Factory Automation and Production Systems (FAPS), Friedrich-Alexander-Universität Erlangen-Nürnberg, Egerlandstr. 7-9, 91058 Erlangen, Germany
| | - Dmitry Volodkin
- Department of Chemistry and Forensics, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom.
| | - Anna Vikulina
- Bavarian Polymer Institute, Friedrich-Alexander-Universität Erlangen-Nürnberg, Dr.-Mack-Straße 77, 90762 Fürth, Germany.
| |
Collapse
|
10
|
Nene LC, Nyokong T. The in-vitro proliferation-suppression of MCF-7 and HeLa cell lines mediated by differently substituted ionic phthalocyanines in sonodynamic therapy supplemented-photodynamic therapy. J Inorg Biochem 2023; 239:112084. [PMID: 36469974 DOI: 10.1016/j.jinorgbio.2022.112084] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022]
Abstract
This work focuses on the study of the effects of the ultrasonic frequency (MHz) and power (W.cm-2) on the stability, reactive oxygen species yields and cytotoxicity activities of differently substituted ionic phthalocyanines (Pcs) in sonodynamic therapy (SDT). Four ultrasonic parameters were investigated: Par I (1 MHz: 1 W.cm-2), Par II (1 MHz: 2 W.cm-2), Par III (3 MHz: 1 W.cm-2) and Par IV (3 MHz: 2 W.cm-2). A higher degradation of the Pcs was observed with increasing power at the Par II. Two reactive oxygen species (ROS) were detected in the ultrasound treated Pcs: singlet oxygen and hydroxyl radicals. Due to minimal degradation of most Pcs, Par I was chosen for SDT, photodynamic therapy (PDT), and photo-sonodynamic therapy (PSDT) against Michigan Cancer Foundation-7 and Henrietta Lacks cancer cell lines. PSDT generally showed improved therapeutic efficacies of the Pcs compared to the SDT and PDT mono treatments.
Collapse
Affiliation(s)
- Lindokuhle Cindy Nene
- Institute of Nanotechnology Innovation, P.O. 94, Rhodes University, Makhanda, South Africa
| | - Tebello Nyokong
- Institute of Nanotechnology Innovation, P.O. 94, Rhodes University, Makhanda, South Africa.
| |
Collapse
|
11
|
He T, Zhang C, Colombani T, Bencherif SA, Porter RM, Bajpayee AG. Intra-articular kinetics of a cartilage targeting cationic PEGylated protein for applications in drug delivery. Osteoarthritis Cartilage 2023; 31:187-198. [PMID: 36241136 PMCID: PMC9892226 DOI: 10.1016/j.joca.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Cartilage targeting cationic glycoprotein Avidin was PEGylated to synthesize a multi-arm Avidin (mAv) nano-construct with high drug loading content. Here we investigate mAv biodistribution and kinetics over a 7-day period following intra-articular (IA) administration in rat knee joints. METHODS Labeled mAv was injected into healthy rat knees, and joint tissues (articular cartilage, menisci, ligaments, tendons, fat pad) were harvested following sacrifice at 6 h, 1, 4 and 7 days. Its IA biodistribution and retention were measured using fluorescence microscopy. Tissue localization was compared in young vs old rats by immunohistochemistry. mAv chondrotoxicity and immune response were evaluated to determine safe carrier dose limits. RESULTS mAv penetrated through the full thickness of rat cartilage and other joint tissues within 6 h, remaining detectable within most joint tissues over 7 days. Intra-tissue uptake correlated strongly with tissue GAG concentration, confirming the dominant role of electrostatic interactions between positively charged mAv and the negatively charged aggrecan proteoglycans. mAv was uptaken by chondrocytes and also penetrated the osteocyte lacuno-canalicular system of peri-articular bone in both young and old rats. mAv did not cause cytotoxicity at concentrations up to 300 μM but elicited a dose dependent immunogenic response. CONCLUSIONS mAv's ability to target a variety of joint tissues, chondrocytes, and peri-articular osteocytes without sequestration in synovial fluid makes it a versatile carrier for delivering a wide range of drugs for treating a broad class of musculoskeletal diseases. Drugs can be conjugated using simple aqueous based avidin-biotin reaction, supporting its clinical prospects.
Collapse
Affiliation(s)
- T He
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - C Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - T Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - S A Bencherif
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - R M Porter
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - A G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA; Department of Mechanical Engineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Nene LC, Buthelezi K, Prinsloo E, Nyokong T. The in vitro photo-sonodynamic combinatorial therapy activity of cationic and zwitterionic phthalocyanines on MCF-7 and HeLa cancer cell lines. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.114116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
15
|
Zhan L, Xiao C, Li C, Zhai J, Yang F, Piao J, Ning C, Zhou Z, Yu P, Qi S. Internal Wireless Electrical Stimulation from Piezoelectric Barium Titanate Nanoparticles as a New Strategy for the Treatment of Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:45032-45041. [PMID: 36153948 DOI: 10.1021/acsami.2c12668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive BC subtype with a higher metastatic rate and a worse 5-year survival ratio than the other BC. It is an urgent need to develop a noninvasive treatment with high efficiency to resist TNBC cell proliferation and invasion. Internal wireless electric stimulation (ES) based on piezoelectric materials is an emerging noninvasive strategy, with adjustable ES intensity and excellent biosafety. In this study, three different barium titanate nanoparticles (BTNPs) with different crystal phases and piezoelectric properties were studied. Varying intensities of internal ES were generated from the three BTNPs (i.e., BTO, U-BTO, P-BTO). In vitro tests revealed that the internal ES from BTNPs was efficient at reducing the proliferative potential of cancer cells, particularly BC cells. In vitro experiments on MDA-MB-231, a typical TNBC cell line, further revealed that the internal wireless ES from BTNPs significantly inhibited cell growth and migration up to about 82% and 60%, respectively. In vivo evaluation of MDA-MB-231 tumor-bearing mice indicated that internal ES not only resisted almost 70% tumor growth but also significantly inhibited lung metastasis. More importantly, in vitro and in vivo studies demonstrated a favorable correlation between the anticancer impact and the intensities of ES. The underlying mechanism of MDA-MB-231 cell proliferation and metastasis inhibition caused by internal ES was also investigated. In summary, our results revealed the effect and mechanism of internal ES from piezoelectric nanoparticles on TNBC cell proliferation and migration regulation and proposed a promising noninvasive therapeutic strategy for TNBC with minimal side effects while exhibiting good therapeutic efficiency.
Collapse
Affiliation(s)
- Lizhen Zhan
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Cairong Xiao
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Changhao Li
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Jinxia Zhai
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Fabang Yang
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Jinhua Piao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Chengyun Ning
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
- China-Singapore International Joint Research Institute, Guangzhou 511365, China
| | - Zhengnan Zhou
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
| | - Peng Yu
- School of Material Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, Metallic Materials Surface Functionalization Engineering Research Center of Guangdong Province, South China University of Technology, Guangzhou 510641, China
- China-Singapore International Joint Research Institute, Guangzhou 511365, China
| | - Suijian Qi
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
16
|
Vedadghavami A, He T, Zhang C, Amiji SM, Hakim B, Bajpayee AG. Charge-based drug delivery to cartilage: Hydrophobic and not electrostatic interactions are the dominant cause of competitive binding of cationic carriers in synovial fluid. Acta Biomater 2022; 151:278-289. [PMID: 35963518 PMCID: PMC10441566 DOI: 10.1016/j.actbio.2022.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/07/2022] [Accepted: 08/05/2022] [Indexed: 01/04/2023]
Abstract
Charge-based drug delivery has proven to be effective for targeting negatively charged cartilage for the treatment of osteoarthritis. Cartilage is surrounded by synovial fluid (SF), which is comprised of negatively charged hyaluronic acid and hydrophobic proteins that can competitively bind cationic carriers and prevent their transport into cartilage. Here we investigate the relative contributions of charge and hydrophobic effects on the binding of cationic carriers within healthy and arthritic SF by comparing the transport of arginine-rich cartilage targeting cationic peptide carriers with hydrophilic (CPC +14N) or hydrophobic property (CPC +14A). CPC +14N had significantly greater intra-cartilage uptake in presence of SF compared to CPC +14A in-vitro and in vivo. In presence of individual anionic SF constituents, both CPCs maintained similar high intra-cartilage uptake while in presence of hydrophobic constituents, CPC +14N had greater uptake confirming that hydrophobic and not charge interactions are the dominant cause of competitive binding within SF. Results also demonstrate that short-range effects can synergistically stabilize intra-cartilage charge-based binding - a property that can be utilized for enhancing drug-carrier residence time in arthritic cartilage with diminished negative fixed charge density. The work provides a framework for the rational design of cationic carriers for developing targeted therapies for another complex negatively charged environments. STATEMENT OF SIGNIFICANCE: This work demonstrates that hydrophobic and not charge interactions are the dominant cause of the binding of cationic carriers in synovial fluid. Therefore, cationic carriers can be effectively used for cartilage targeting if they are made hydrophilic. This can facilitate clinical translation of various osteoarthritis drugs for cartilage repair that have failed due to a lack of effective cartilage targeting methods. It also demonstrates that short-range hydrogen bonds can synergistically stabilize electrostatic binding in cartilage offering a method for enhancing the targeting and residence time of cationic carriers within arthritic cartilage with reduced charge density. Finally, the cartilage-synovial fluid unit provides an excellent model of a complex negatively charged environment and allows us to generalize these findings and develop targeted therapies for other charged tissue-systems.
Collapse
Affiliation(s)
- Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Tengfei He
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Salima M Amiji
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Bill Hakim
- 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
|
17
|
Parrasia S, Szabò I, Zoratti M, Biasutto L. Peptides as Pharmacological Carriers to the Brain: Promises, Shortcomings and Challenges. Mol Pharm 2022; 19:3700-3729. [PMID: 36174227 DOI: 10.1021/acs.molpharmaceut.2c00523] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Central nervous system (CNS) diseases are among the most difficult to treat, mainly because the vast majority of the drugs fail to cross the blood-brain barrier (BBB) or to reach the brain at concentrations adequate to exert a pharmacological activity. The obstacle posed by the BBB has led to the in-depth study of strategies allowing the brain delivery of CNS-active drugs. Among the most promising strategies is the use of peptides addressed to the BBB. Peptides are versatile molecules that can be used to decorate nanoparticles or can be conjugated to drugs, with either a stable link or as pro-drugs. They have been used to deliver to the brain both small molecules and proteins, with applications in diverse therapeutic areas such as brain cancers, neurodegenerative diseases and imaging. Peptides can be generally classified as receptor-targeted, recognizing membrane proteins expressed by the BBB microvessels (e.g., Angiopep2, CDX, and iRGD), "cell-penetrating peptides" (CPPs; e.g. TAT47-57, SynB1/3, and Penetratin), undergoing transcytosis through unspecific mechanisms, or those exploiting a mixed approach. The advantages of peptides have been extensively pointed out, but so far few studies have focused on the potential negative aspects. Indeed, despite having a generally good safety profile, some peptide conjugates may display toxicological characteristics distinct from those of the peptide itself, causing for instance antigenicity, cardiovascular alterations or hemolysis. Other shortcomings are the often brief lifetime in vivo, caused by the presence of peptidases, the vulnerability to endosomal/lysosomal degradation, and the frequently still insufficient attainable increase of brain drug levels, which remain below the therapeutically useful concentrations. The aim of this review is to analyze not only the successful and promising aspects of the use of peptides in brain targeting but also the problems posed by this strategy for drug delivery.
Collapse
Affiliation(s)
- Sofia Parrasia
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| |
Collapse
|
18
|
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
|
19
|
Dash S, Zuo J, Steyger PS. Local Delivery of Therapeutics to the Cochlea Using Nanoparticles and Other Biomaterials. Pharmaceuticals (Basel) 2022; 15:1115. [PMID: 36145336 PMCID: PMC9504900 DOI: 10.3390/ph15091115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022] Open
Abstract
Hearing loss negatively impacts the well-being of millions of people worldwide. Systemic delivery of ototherapeutics has limited efficacy due to severe systemic side effects and the presence of the blood-labyrinth barrier that selectively limits or enables transfer of molecules between plasma and inner ear tissues and fluids. Local drug delivery into the middle and inner ear would be preferable for many newly emerging classes of drugs. Although the cochlea is a challenging target for drug delivery, recent technologies could provide a safe and efficacious delivery of ototherapeutics. Local drug delivery routes include topical delivery via the external auditory meatus, retroauricular, transtympanic, and intracochlear delivery. Many new drug delivery systems specifically for the inner ear are under development or undergoing clinical studies. Future studies into these systems may provide a means for extended delivery of drugs to preserve or restore hearing in patients with hearing disorders. This review outlines the anatomy of the (inner) ear, describes the various local delivery systems and routes, and various quantification methodologies to determine the pharmacokinetics of the drugs in the inner ear.
Collapse
Affiliation(s)
| | | | - Peter S. Steyger
- Translational Hearing Center, Department of Biomedical Sciences, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA
| |
Collapse
|
20
|
Vedadghavami A, Hakim B, He T, Bajpayee AG. Cationic peptide carriers enable long-term delivery of insulin-like growth factor-1 to suppress osteoarthritis-induced matrix degradation. Arthritis Res Ther 2022; 24:172. [PMID: 35858920 PMCID: PMC9297664 DOI: 10.1186/s13075-022-02855-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Insulin-like growth factor-1 (IGF-1) has the potential to be used for osteoarthritis (OA) treatment but has not been evaluated in clinics yet owing to toxicity concerns. It suffers from short intra-joint residence time and a lack of cartilage targeting following its intra-articular administration. Here, we synthesize an electrically charged cationic formulation of IGF-1 by using a short-length arginine-rich, hydrophilic cationic peptide carrier (CPC) with a net charge of +14, designed for rapid and high uptake and retention in both healthy and arthritic cartilage. METHODS IGF-1 was conjugated to CPC by using a site-specific sulfhydryl reaction via a bifunctional linker. Intra-cartilage depth of penetration and retention of CPC-IGF-1 was compared with the unmodified IGF-1. The therapeutic effectiveness of a single dose of CPC-IGF-1 was compared with free IGF-1 in an IL-1α-challenged cartilage explant culture post-traumatic OA model. RESULTS CPC-IGF-1 rapidly penetrated through the full thickness of cartilage creating a drug depot owing to electrostatic interactions with negatively charged aggrecan-glycosaminoglycans (GAGs). CPC-IGF-1 remained bound within the tissue while unmodified IGF-1 cleared out. Treatment with a single dose of CPC-IGF-1 effectively suppressed IL-1α-induced GAG loss and nitrite release and rescued cell metabolism and viability throughout the 16-day culture period, while free IGF at the equivalent dose was not effective. CONCLUSIONS CPC-mediated depot delivery of IGF-1 protected cartilage by suppressing cytokine-induced catabolism with only a single dose. CPC is a versatile cationic motif that can be used for intra-cartilage delivery of other similar-sized drugs.
Collapse
Affiliation(s)
| | - Bill Hakim
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Tengfei He
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Departments of Mechanical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
21
|
Young CC, Byrne JD, Wentworth AJ, Collins JE, Chu JN, Traverso G. Respirators in Healthcare: Material, Design, Regulatory, Environmental, and Economic Considerations for Clinical Efficacy. GLOBAL CHALLENGES (HOBOKEN, NJ) 2022; 6:2200001. [PMID: 35601599 PMCID: PMC9110919 DOI: 10.1002/gch2.202200001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Indexed: 06/15/2023]
Abstract
Maintaining an ample supply of personal protective equipment continues to be a challenge for the healthcare industry, especially during emergency situations and times of strain on the supply chain. Most critically, healthcare workers exposed to potential airborne hazards require sufficient respiratory protection. Respirators are the only type of personal protective equipment able to provide adequate respiratory protection. However, their ability to shield hazards depends on design, material, proper fit, and environmental conditions. As a result, not all respirators may be adequate for all scenarios. Additionally, factors including user comfort, ease of use, and cost contribute to respirator effectiveness. Therefore, a careful consideration of these parameters is essential for ensuring respiratory protection for those working in the healthcare industry. Here respirator design and material characteristics are reviewed, as well as properties of airborne hazards and potential filtration mechanisms, regulatory standards of governmental agencies, respirator efficacy in the clinical setting, attitude of healthcare personnel toward respiratory protection, and environmental and economic considerations of respirator manufacturing and distribution.
Collapse
Affiliation(s)
- Cameron C. Young
- Division of GastroenterologyBrigham and Women's HospitalHarvard Medical School75 Francis StBostonMA02115USA
- Departments of Chemical Engineering and BiochemistryNortheastern University300 Huntington AveBostonMA02115USA
| | - James D. Byrne
- Division of GastroenterologyBrigham and Women's HospitalHarvard Medical School75 Francis StBostonMA02115USA
- Harvard Radiation Oncology Program55 Fruit StBostonMA02114USA
- David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology500 Main St. Building 76CambridgeMA02142USA
- Department of Mechanical EngineeringMassachusetts Institute of Technology77 Massachusetts AveCambridgeMA02139USA
- Department of Radiation OncologyDana‐Farber Cancer Institute/Brigham and Women's Hospital44 Binney StBostonMA02115USA
| | - Adam J. Wentworth
- Division of GastroenterologyBrigham and Women's HospitalHarvard Medical School75 Francis StBostonMA02115USA
- David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology500 Main St. Building 76CambridgeMA02142USA
- Department of Mechanical EngineeringMassachusetts Institute of Technology77 Massachusetts AveCambridgeMA02139USA
| | - Joy E. Collins
- David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology500 Main St. Building 76CambridgeMA02142USA
- Department of Mechanical EngineeringMassachusetts Institute of Technology77 Massachusetts AveCambridgeMA02139USA
- Division of GastroenterologyMassachusetts General Hospital55 Fruit StBostonMA02114USA
| | - Jacqueline N. Chu
- David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology500 Main St. Building 76CambridgeMA02142USA
| | - Giovanni Traverso
- Division of GastroenterologyBrigham and Women's HospitalHarvard Medical School75 Francis StBostonMA02115USA
- David H. Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology500 Main St. Building 76CambridgeMA02142USA
- Department of Mechanical EngineeringMassachusetts Institute of Technology77 Massachusetts AveCambridgeMA02139USA
| |
Collapse
|
22
|
He T, Shaw I, Vedadghavami A, Bajpayee AG. Single-Dose Intra-Cartilage Delivery of Kartogenin Using a Cationic Multi-Arm Avidin Nanocarrier Suppresses Cytokine-Induced Osteoarthritis-Related Catabolism. Cartilage 2022; 13:19476035221093072. [PMID: 35491681 PMCID: PMC9251829 DOI: 10.1177/19476035221093072] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/01/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Kartogenin (KGN) has proven as a both chondrogenic and chondroprotective drug for osteoarthritis (OA) therapy. However, being a small hydrophobic molecule, KGN suffers from rapid joint clearance and inability to penetrate cartilage to reach chondrocytes following intra-articular administration. As such multiple high doses are needed that can lead to off-target effects including stimulation and tissue outgrowth. Here we design charge-based cartilage targeting formulation of KGN by using a multi-arm cationic nano-construct of Avidin (mAv) that can rapidly penetrate into cartilage in high concentrations owing to weak-reversible electrostatic binding interactions with negatively charged aggrecan-glycosaminoglycans (GAGs) and form an extended-release drug depot such that its therapeutic benefit can be reaped in just a single dose. DESIGN We synthesized 2 novel formulations, one with a releasable ester linker (mAv-OH-KGN, release half-life ~58 h) that enables sustained KGN release over 2 weeks and another with a non-releasable amide linker (mAv-NH-KGN) that relies on mAv's ability to be uptaken and endocytosed by chondrocytes for drug delivery. Their effectiveness in suppressing cytokine-induced catabolism was evaluated in vitro using cartilage explant culture model. RESULTS A single 100 μM dose of cartilage homing mAv-KGN was significantly more effective in suppressing cytokine-induced GAG loss, cell death, inflammatory response and in rescuing cell metabolism than a single dose of free KGN; multiple doses of free KGN were needed to match this therapeutic response. CONCLUSION mAv mediated delivery of KGN is promising and can facilitate clinical translation of KGN for OA treatment with only a single dose.
Collapse
Affiliation(s)
- Tengfei He
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
| | - Irfhan Shaw
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
| | | | - Ambika G. Bajpayee
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
- Department of Mechanical Engineering,
Northeastern University, Boston, MA, USA
| |
Collapse
|
23
|
Activatable Peptides for Rapid and Simple Visualization of Protease Activity Secreted in Living Cells. Int J Mol Sci 2022; 23:ijms23031605. [PMID: 35163529 PMCID: PMC8836073 DOI: 10.3390/ijms23031605] [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: 12/24/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 11/30/2022] Open
Abstract
Activity-based monitoring of cell-secreted proteases has gained significant interest due to the implication of these substances in diverse cellular functions. Here, we demonstrated a cell-based method of monitoring protease activity using fluorescent cell-permeable peptides. The activatable peptide consists of anionic (EEEE), cleavable, and cationic sequences (RRRR) that enable intracellular delivery by matrix metalloproteinase-2 (MMP2), which is secreted by living cancer cells. Compared to HT-29 cells (MMP2-negative), HT-1080 cells (MMP2-positive) showed a strong fluorescence response to the short fluorescent peptide via cell-secreted protease activation. Our approach is expected to find applications for the rapid visualization of protease activity in living cells.
Collapse
|
24
|
Iqbal S, Zhao Z. Poly (β amino esters) copolymers: Novel potential vectors for delivery of genes and related therapeutics. Int J Pharm 2022; 611:121289. [PMID: 34775041 DOI: 10.1016/j.ijpharm.2021.121289] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022]
Abstract
The unique properties of polymers have performed an essential contribution to the drug delivery system by providing an outstanding platform for the delivery of macromolecules and genes. However, the block copolymers have been the subject of many recently published works whose results have demonstrated excellent performance in drug targeting. Poly(β-amino esters) (PβAEs) copolymers are the synthetic cationic polymers that are tailored by chemically joining PβAEs with other additives to demonstrate extraordinary efficiency in designing pre-defined and pre-programmed nanostructures, site-specific delivery, andovercoming the distinct cellular barriers. Different compositional and structural libraries could be generated by combinatorial chemistry and by the addition of various novel functional additives that fulfill the multiple requirements of targeted delivery. These intriguing attributes allow PβAE-copolymers to have customized therapeutic functions such as excellent encapsulation capacity, high stability, and stimuli-responsive release. Here, we give an overview of PβAE copolymers-based formulations along with focusing on most notable improvements such as structural modifications, bio-conjugations, and stimuli-responsive approaches, for safe and effective nucleic acids delivery.
Collapse
Affiliation(s)
- Sajid Iqbal
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhongxi Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Pediatric Pharmaceutical Engineering Laboratory of Shandong Province, Shandong Dyne Marine Biopharmaceutical Company Limited, Rongcheng, Shandong 264300, PR China; Chemical Immunopharmaceutical Engineering Laboratory of Shandong Province, Shandong Xili Pharmaceutical Company Limited, Heze, Shandong 274300, PR China.
| |
Collapse
|
25
|
Mehta S, Young CC, Warren MR, Akhtar S, Shefelbine SJ, Crane JD, Bajpayee AG. Resveratrol and Curcumin Attenuate Ex Vivo Sugar-Induced Cartilage Glycation, Stiffening, Senescence, and Degeneration. Cartilage 2021; 13:1214S-1228S. [PMID: 33472415 PMCID: PMC8804818 DOI: 10.1177/1947603520988768] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Advanced glycation end-product (AGE) accumulation is implicated in osteoarthritis (OA) pathogenesis in aging and diabetic populations. Here, we develop a representative nonenzymatic glycation-induced OA cartilage explant culture model and investigate the effectiveness of resveratrol, curcumin, and eugenol in inhibiting AGEs and the structural and biological hallmarks of cartilage degeneration. DESIGN Bovine cartilage explants were treated with AGE-bovine serum albumin, threose, and ribose to determine the optimal conditions that induce physiological levels of AGEs while maintaining chondrocyte viability. AGE crosslinks, tissue stiffness, cell viability, metabolism and senescence, nitrite release and loss of glycosaminoglycans were assessed. Explants were cotreated with resveratrol, curcumin, or eugenol to evaluate their anti-AGE properties. Blind docking analysis was conducted to estimate binding energies of drugs with collagen II. RESULTS Treatment with 100 mM ribose significantly increased AGE crosslink formation and tissue stiffness, resulting in reduced chondrocyte metabolism and enhanced senescence. Blind docking analysis revealed stronger binding energies of both resveratrol and curcumin than ribose, with glycation sites along a human collagen II fragment, indicating their increased likelihood of competitively inhibiting ribose activity. Resveratrol and curcumin, but not eugenol, successfully inhibited AGE crosslink formation and its associated downstream biological response. CONCLUSIONS We establish a cartilage explant model of OA that recapitulates several aspects of aged human cartilage. We find that resveratrol and curcumin are effective anti-AGE therapeutics with the potential to decelerate age-related and diabetes-induced OA. This in vitro nonenzymatic glycation-induced model provides a tool for screening OA drugs, to simultaneously evaluate AGE-induced biological and mechanical changes.
Collapse
Affiliation(s)
- Shikhar Mehta
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
| | - Cameron C. Young
- Department of Chemical Engineering,
Northeastern University, Boston, MA, USA
| | - Matthew R. Warren
- Department of Bioengineering,
Northeastern University, Boston, MA, USA
| | - Sumayyah Akhtar
- Department of Biochemistry, Northeastern
University, Boston, MA, USA
| | - Sandra J. Shefelbine
- Department of Bioengineering,
Northeastern University, Boston, MA, USA,Department of Mechanical &
Industrial Engineering, Northeastern University, Boston, MA, USA
| | - Justin D. Crane
- Department of Biology, Northeastern
University, Boston, MA, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering,
Northeastern University, Boston, MA, USA,Department of Mechanical &
Industrial Engineering, Northeastern University, Boston, MA, USA,Ambika G. Bajpayee, Department of
Bioengineering, Northeastern University, ISEC Room 216, 805 Columbus Avenue,
Boston, MA 02115, USA.
| |
Collapse
|
26
|
Warren MR, Zhang C, Vedadghavami A, Bokvist K, Dhal PK, Bajpayee AG. Milk exosomes with enhanced mucus penetrability for oral delivery of siRNA. Biomater Sci 2021; 9:4260-4277. [PMID: 33367332 PMCID: PMC8205963 DOI: 10.1039/d0bm01497d] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bovine milk-derived exosomes have recently emerged as a promising nano-vehicle for the encapsulation and delivery of macromolecular biotherapeutics. Here we engineer high purity bovine milk exosomes (mExo) with modular surface tunability for oral delivery of small interfering RNA (siRNA). We utilize a low-cost enrichment method combining casein chelation with differential ultracentrifugation followed by size exclusion chromatography, yielding mExo of high concentration and purity. Using in vitro models, we demonstrate that negatively charged hydrophobic mExos can penetrate multiple biological barriers to oral drug delivery. A hydrophilic polyethylene glycol (PEG) coating was introduced on the mExo surface via passive, stable hydrophobic insertion of a conjugated lipid tail, which significantly reduced mExo degradation in acidic gastric environment and enhanced their permeability through mucin by over 3× compared to unmodified mExo. Both mExo and PEG-mExo exhibited high uptake by intestinal epithelial cells and mediated functional intracellular delivery of siRNA, thereby suppressing the expression of the target green fluorescence protein (GFP) gene by up to 70%. We also show that cationic chemical transfection is significantly more efficient in loading siRNA into mExo than electroporation. The simplicity of isolating high purity mExo in high concentrations and equipping them with tunable surface properties, demonstrated here, paves way for the development of mExo as an effective, scalable platform technology for oral drug delivery of siRNA.
Collapse
Affiliation(s)
- Matthew R Warren
- 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.
| | | | | | - Ambika G Bajpayee
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA. and Mechanical Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Siefen T, Lokhnauth J, Liang A, Larsen CC, Lamprecht A. An ex-vivo model for transsynovial drug permeation of intraarticular injectables in naive and arthritic synovium. J Control Release 2021; 332:581-591. [PMID: 33705826 DOI: 10.1016/j.jconrel.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/23/2021] [Accepted: 03/04/2021] [Indexed: 01/17/2023]
Abstract
Estimation of joint residence time of a drug is a key requirement for rational development of intraarticular therapeutics. There is a great need for a predictive model to reduce the high number of animal experiments in early stage development. Here, a Franz-cell based porcine ex-vivo permeation model is proposed, and transsynovial permeation of fluorescently-labeled dextrans in the range of potential drug candidates (10-150 kDa), as well as a small molecule (fluorescein sodium) and charged dextran derivates, have been determined. In addition, a lipopolysaccharide (LPS) -induced synovitis model was assessed for inflammatory biomarker levels and its effect on permeation of the solutes. Size-dependent permeability was observed for the analytes, which distinctly differed from findings with an artificial polycarbonate membrane, which is a widely used model. LPS was found to successfully stimulate an inflammatory response and led to a reduced size selectivity of the synovial membrane. 150 kDa dextran flux was accelerated approximately 2.5-fold in the inflamed state, whereas the permeation of smaller molecules was little affected. Moreover, by varying the LPS concentrations, the ex-vivo model was shown to produce varying degrees of synovitis-like inflammation. A simple and highly relevant ex-vivo tool for investigation of transsynovial permeation was developed, offering the further advantage of mimicking synovitis-induced permeability changes. Thus, this model provides a promising method for formulation screening, while reducing the need for animal experiments.
Collapse
Affiliation(s)
- Tobias Siefen
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | | | - Alfred Liang
- Ferring Pharmaceuticals Inc, Parsippany, NJ, USA
| | | | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; PEPITE (EA4267), University of Burgundy/Franche-Comté, Besançon, France.
| |
Collapse
|
28
|
Gawthrop PJ, Pan M. Network Thermodynamical Modeling of Bioelectrical Systems: A Bond Graph Approach. Bioelectricity 2021; 3:3-13. [PMID: 34476374 DOI: 10.1089/bioe.2020.0042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interactions among biomolecules, electrons, and protons are essential to many fundamental processes sustaining life. It is therefore of interest to build mathematical models of these bioelectrical processes not only to enhance understanding but also to enable computer models to complement in vitro and in vivo experiments. Such models can never be entirely accurate; it is nevertheless important that the models are compatible with physical principles. Network Thermodynamics, as implemented with bond graphs, provide one approach to creating physically compatible mathematical models of bioelectrical systems. This is illustrated using simple models of ion channels, redox reactions, proton pumps, and electrogenic membrane transporters thus demonstrating that the approach can be used to build mathematical and computer models of a wide range of bioelectrical systems.
Collapse
Affiliation(s)
- Peter J Gawthrop
- Systems Biology Laboratory, Department of Biomedical Engineering, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia.,Systems Biology Laboratory, School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| | - Michael Pan
- Systems Biology Laboratory, Department of Biomedical Engineering, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia.,Systems Biology Laboratory, School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
29
|
He T, Li B, Colombani T, Joshi-Navare K, Mehta S, Kisiday J, Bencherif SA, Bajpayee AG. Hyaluronic Acid-Based Shape-Memory Cryogel Scaffolds for Focal Cartilage Defect Repair. Tissue Eng Part A 2021; 27:748-760. [PMID: 33108972 DOI: 10.1089/ten.tea.2020.0264] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Traumatic joint injuries can result in significant cartilage defects, which can greatly increase the risk of osteoarthritis development. Due to the limited self-healing capacity of avascular cartilage, tissue engineering approaches are required for filling defects and promoting cartilage regeneration. Current approaches utilize invasive surgical procedures for extraction and implantation of autologous chondrocytes; therefore, injectable biomaterials have gained interest to minimize the risk of infection as well as patient pain and discomfort. In this study, we engineered biomimetic, hyaluronic acid (HA)-based cryogel scaffolds that possess shape-memory properties as they contract and regain their shape after syringe injection to noninvasively fill cartilage defects. The cryogels, fabricated with HA and glycidyl methacrylate at -20°C, resulted in an elastic, macroporous, and highly interconnected network that provided a conducive microenvironment for chondrocytes to remain viable and metabolically active after injection through a syringe needle. Chondrocytes seeded within cryogels and cultured for 15 days exhibited enhanced cell proliferation, metabolism, and production of cartilage extracellular matrix glycosaminoglycans compared with HA-based hydrogels. Furthermore, immunohistochemical staining revealed production of collagen type II from chondrocyte-seeded cryogels, indicating the maintenance of cell phenotype. These results demonstrate the potential of chondrocyte-seeded, HA-based, injectable cryogel scaffolds to promote regeneration of cartilage tissue for nonsurgically invasive defect repair. Impact statement Hyaluronic acid-based shape-memory cryogels provide a conducive microenvironment for chondrocyte adhesion, proliferation, and matrix biosynthesis for use in repair of cartilage defects. Due to their sponge-like elastic properties, cryogels can fully recover their original shape back after injection while not impacting metabolism or viability of encapsulated cells. Clinically, they provide an opportunity for filling focal cartilage defects by using a single, minimally invasive injection of a cell encapsulating biocompatible three-dimensional scaffold that can return to its original structure to fit the defect geometry and enable matrix regeneration.
Collapse
Affiliation(s)
- Tengfei He
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - Boting Li
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Kasturi Joshi-Navare
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Shikhar Mehta
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA
| | - John Kisiday
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sidi A Bencherif
- Department of Bioengineering and Northeastern University, Boston, Massachusetts, USA.,Department of Chemical Engineering, 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
|
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
|
Yadav KS, Upadhya A, Misra A. Targeted drug therapy in nonsmall cell lung cancer: clinical significance and possible solutions-part II (role of nanocarriers). Expert Opin Drug Deliv 2020; 18:103-118. [PMID: 33017541 DOI: 10.1080/17425247.2021.1832989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Nonsmall cell lung cancer (NSCLC) accounts for 80-85% of the cases of lung cancer. The conventional therapeutic effective dosage forms used to treat NSCLC are associated with rigid administration schedules, adverse effects, and may be associated with acquired resistance to therapy. Nanocarriers may provide a suitable alternative to regular formulations to overcome inherent drawbacks and provide better treatment modalities for the patient. AREAS COVERED The article explores the application of drug loaded nanocarriers for lung cancer treatment. Drug-loaded nanocarriers can be modified to achieve controlled delivery at the desired tumor infested site. The type of nanocarriers employed are diverse based on polymers, liposomes, metals and a combination of two or more different base materials (hybrids). These may be designed for systemic delivery or local delivery to the lung compartment (via inhalation). EXPERT OPINION Nanocarriers can improve pharmacokinetics of the drug payload by improving its delivery to the desired location and can reduce associated systemic toxicities. Through nanocarriers, a wide variety of therapeutics can be administered and targeted to the cancerous site. Some examples of the utilities of nanocarriers are codelivery of drugs, gene delivery, and delivery of other biologics. Overall, the nanocarriers have promising potential in improving therapeutic efficacy of drugs used in NSCLC.
Collapse
Affiliation(s)
- Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Svkm's Nmims , Mumbai, India
| | - Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Svkm's Nmims , Mumbai, India
| | - Ambikanandan Misra
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Svkm's Nmims , Mumbai, India
| |
Collapse
|
32
|
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
|
33
|
Wagner EK, Vedadghavami A, Jacobsen TD, Goel SA, Chahine NO, Bajpayee AG. Avidin grafted dextran nanostructure enables a month-long intra-discal retention. Sci Rep 2020; 10:12017. [PMID: 32694557 PMCID: PMC7374582 DOI: 10.1038/s41598-020-68351-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
Low back pain is often the direct result of degeneration of the intervertebral disc. A wide range of therapeutics including anti-catabolic, pro-anabolic factors and chemo-attractants that can stimulate resident cells and recruit endogenous progenitors are under consideration. The avascular nature and the dense matrix of this tissue make it challenging for systemically administered drugs to reach their target cells inside the nucleus pulposus (NP), the central gelatinous region of the intervertebral disc (IVD). Therefore, local intra-discal injection of therapeutic drugs directly into the NP is a clinically relevant delivery approach, however, suffers from rapid and wide diffusion outside the injection site resulting in short lived benefits while causing systemic toxicity. NP has a high negative fixed charge density due to the presence of negatively charged aggrecan glycosaminoglycans that provide swelling pressures, compressive stiffness and hydration to the tissue. This negative fixed charge density can also be used for enhancing intra-NP residence time of therapeutic drugs. Here we design positively charged Avidin grafted branched Dextran nanostructures that utilize long-range binding effects of electrostatic interactions to bind with the intra-NP negatively charged groups. The binding is strong enough to enable a month-long retention of cationic nanostructures within the NP following intra-discal administration, yet weak and reversible to allow movement to reach cells dispersed throughout the tissue. The branched carrier has multiple sites for drug conjugation and can reduce the need for multiple injections of high drug doses and minimize associated side-effects, paving the way for effective clinical translation of potential therapeutics for treatment of low back pain and disc degeneration.
Collapse
Affiliation(s)
- Erica K Wagner
- Department of Bioengineering, Northeastern University, 805 Columbus Avenue, Boston, MA, 02120, USA
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, 805 Columbus Avenue, Boston, MA, 02120, USA
| | - Timothy D Jacobsen
- Department of Orthopedic Surgery, Columbia University, 650 West 168th Street, 14-1410, New York, NY, 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Shakti A Goel
- Department of Orthopedic Surgery, Indian Spinal Injuries Center, New Delhi, India
| | - Nadeen O Chahine
- Department of Orthopedic Surgery, Columbia University, 650 West 168th Street, 14-1410, New York, NY, 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, 805 Columbus Avenue, Boston, MA, 02120, USA.
- Department of Mechanical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|