Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mater Chem B ; 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38946491

RESUMO

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.

2.
Small Methods ; : e2301443, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38607953

RESUMO

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.

3.
Biomater Sci ; 12(3): 634-649, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38047368

RESUMO

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.


Assuntos
Exossomos , Nanopartículas , Animais , Suínos , Leite , Exossomos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Peptídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Permeabilidade , Muco/metabolismo , Administração Oral , Portadores de Fármacos/química , Nanopartículas/química
4.
ACS Nano ; 17(7): 6649-6663, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36989423

RESUMO

Cartilage tissue exhibits early degenerative changes with onset of osteoarthritis (OA). Early diagnosis is critical as there is only a narrow time window during which therapeutic intervention can reverse disease progression. Computed tomography (CT) has been considered for cartilage imaging as a tool for early OA diagnosis by introducing radio-opaque contrast agents like ioxaglate (IOX) into the joint. IOX, however, is anionic and thus repelled by negatively charged cartilage glycosaminoglycans (GAGs) that hinders its intra-tissue penetration and partitioning, resulting in poor CT attenuation. This is further complicated by its short intra-tissue residence time owing to rapid clearance from joints, which necessitates high doses causing toxicity concerns. Here we engineer optimally charged cationic contrast agents based on cartilage negative fixed charge density by conjugating cartilage targeting a cationic peptide carrier (CPC) and multi-arm avidin nanoconstruct (mAv) to IOX, such that they can penetrate through the full thickness of cartilage within 6 h using electrostatic interactions and elicit similar CT signal with about 40× lower dose compared to anionic IOX. Their partitioning and distribution correlate strongly with spatial GAG distribution within healthy and early- to late-stage arthritic bovine cartilage tissues at 50-100× lower doses than other cationic contrast agents used in the current literature. The use of contrast agents at low concentrations also allowed for delineation of cartilage from subchondral bone as well as other soft tissues in rat tibial joints. These contrast agents are safe to use at current doses, making CT a viable imaging modality for early detection of OA and staging of its severity.


Assuntos
Cartilagem Articular , Osteoartrite , Ratos , Animais , Bovinos , Meios de Contraste/uso terapêutico , Cartilagem Articular/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Ácido Ioxáglico/uso terapêutico , Cátions , Osteoartrite/diagnóstico por imagem , Diagnóstico Precoce
5.
J Orthop Res ; 41(2): 386-395, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35578981

RESUMO

Adolescent obesity has risen dramatically in the last few decades. While adult obesity may be osteoprotective, the effects of obesity during adolescence, which is a period of massive bone accrual, are not clear. We used a murine model of induced adolescent obesity to examine the structural, mechanical, and compositional differences between obese and healthy weight bone in 16-week-old female C57Bl6 mice. We also examined the effects of a return to normal weight after skeletal maturity (24 weeks old). We found obese adolescent bone exhibited decreased trabecular bone volume, increased cortical diameter, increased ultimate stress, and increased brittleness (decreased plastic energy to fracture), similar to an aging phenotype. The trabecular bone deficits remained after return to normal weight after skeletal maturity. However, after returning to normal diet, there was no difference in ultimate stress nor plastic energy to fracture between groups as the normal diet group increased ultimate stress and brittleness. Interestingly, compositional changes appeared in the former high-fat diet mice after skeletal maturity with a lower mineral to matrix ratio compared to normal diet mice. In addition there was a trend toward increased fluorescent advanced glycation endproducts in the former high-fat diet mice compared to normal diet mice but this did not reach significance (p < 0.05) due to the large variability. The skeletal consequences of adolescent obesity may have lasting implications for the adult skeleton even after return to normal weight. Given the rates of adolescent obesity, skeletal health should be a concern.


Assuntos
Fraturas Ósseas , Obesidade Infantil , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osso e Ossos , Osso Esponjoso , Dieta Hiperlipídica/efeitos adversos , Densidade Óssea
6.
Acta Biomater ; 151: 278-289, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35963518

RESUMO

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.


Assuntos
Cartilagem Articular , Osteoartrite , Arginina/farmacologia , Ligação Competitiva , Cartilagem/metabolismo , Cartilagem Articular/metabolismo , Cátions/química , Portadores de Fármacos/química , Humanos , Ácido Hialurônico/farmacologia , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Peptídeos/química , Líquido Sinovial/metabolismo
7.
Arthritis Res Ther ; 24(1): 172, 2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35858920

RESUMO

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.


Assuntos
Cartilagem Articular , Osteoartrite , Cartilagem Articular/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like I/uso terapêutico , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Peptídeos/farmacologia
8.
Biophys J ; 121(18): 3542-3561, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35765244

RESUMO

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.


Assuntos
Cartilagem Articular , Portadores de Fármacos , Animais , Arginina , Cátions/química , Bovinos , Portadores de Fármacos/química , Glicosaminoglicanos , Modelos Biológicos , Peptídeos/química
9.
Cartilage ; 13(2): 19476035221093072, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35491681

RESUMO

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.


Assuntos
Avidina , Osteoartrite , Anilidas , Avidina/farmacologia , Avidina/uso terapêutico , Cartilagem , Citocinas , Humanos , Nanoestruturas , Osteoartrite/tratamento farmacológico , Ácidos Ftálicos
10.
Methods Mol Biol ; 2394: 537-553, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35094345

RESUMO

Here we describe methods for synthesizing cationic multiarm Avidin (mAv) nanoconstruct that has a wide range of applications in drug delivery and imaging for a variety of negatively charged tissues. The multiarm structure provides multiple sites for covalent conjugation of drugs. We use avidin-biotin reaction that gives the flexibility for conjugating any desired biotinylated drug to mAv by simple mixing at room temperature. We also describe methods to control hydrolysis rates of ester linkers to enable sustained (and tunable) drug release rates in therapeutic doses.


Assuntos
Avidina , Sistemas de Liberação de Medicamentos , Avidina/química , Biotina , Cátions , Tecnologia
11.
Methods Mol Biol ; 2393: 797-812, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34837212

RESUMO

Here we describe methods for synthesizing cationic contrast agents for computed tomography (CT) of cartilage for early diagnosis of tissue degeneration. CT imaging of soft tissues like cartilage is possible only if radio-opaque contrast agents (e.g., ioxaglate) can penetrate through the full thickness of tissue in sufficient concentrations. Ioxaglate (IOX), however, is anionic and is repelled by the negatively charged cartilage matrix resulting in poor CT attenuation. Here we demonstrate cartilage penetrating cationic contrast agents using multi-arm Avidin (mAv) conjugated to ioxaglate (mAv-IOX). mAv-IOX rapidly penetrates through the full thickness of cartilage in high concentrations owing to weak-reversible nature of electrostatic interactions resulting in high CT attenuation even with low doses unlike IOX. The technology has the potential for enabling clinical CT of cartilage and other negatively charged soft tissues.


Assuntos
Osteoartrite , Avidina , Cartilagem Articular/diagnóstico por imagem , Cátions , Meios de Contraste , Diagnóstico Precoce , Humanos , Ácido Ioxáglico , Osteoartrite/diagnóstico por imagem , Tomografia Computadorizada por Raios X
12.
Bioelectricity ; 4(4): 248-258, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36644714

RESUMO

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.

13.
Cartilage ; 13(2_suppl): 1214S-1228S, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33472415

RESUMO

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.


Assuntos
Curcumina , Idoso , Cartilagem/metabolismo , Curcumina/farmacologia , Produtos Finais de Glicação Avançada/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , Humanos , Resveratrol/metabolismo , Resveratrol/farmacologia , Açúcares/metabolismo , Açúcares/farmacologia
14.
Biomater Sci ; 9(12): 4260-4277, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33367332

RESUMO

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.


Assuntos
Exossomos , Animais , Bovinos , Sistemas de Liberação de Medicamentos , Leite , Muco , Polietilenoglicóis , RNA Interferente Pequeno
15.
Curr Opin Rheumatol ; 33(1): 94-109, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33229973

RESUMO

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.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Osteoartrite/tratamento farmacológico , Agrecanas/administração & dosagem , Animais , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Matriz Extracelular/metabolismo , Terapia Genética/métodos , Humanos , Inflamação/tratamento farmacológico , Injeções Intra-Articulares/métodos , Nanopartículas/administração & dosagem , Dor/tratamento farmacológico , Esteroides/administração & dosagem , Membrana Sinovial/metabolismo
16.
Tissue Eng Part A ; 27(11-12): 748-760, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33108972

RESUMO

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.


Assuntos
Cartilagem Articular , Criogéis , Cartilagem , Condrócitos , Humanos , Ácido Hialurônico/farmacologia , Porosidade , Engenharia Tecidual , Alicerces Teciduais
17.
Nano Today ; 342020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32802145

RESUMO

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.

18.
Bioelectricity ; 2(2): 68-81, 2020 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-32803148

RESUMO

Biological systems overwhelmingly comprise charged entities generating electrical activity that can have significant impact on biological structure and function. This intrinsic bio-electrical activity can also be harnessed for overcoming the tissue matrix and cell membrane barriers, which have been outstanding challenges for targeted drug delivery, by using rationally designed cationic carriers. The weak and reversible long-range electrostatic interactions with fixed negatively charged groups facilitate electro-diffusive transport of cationic therapeutics through full-tissue thickness to effectively reach intra-tissue, cellular, and intracellular target sites. This article presents a perspective on the promise of using rationally designed cationic biomaterials in targeted drug delivery, the underlying charge-based mechanisms, and bio-transport phenomena while addressing outstanding concerns around toxicity and methods to mitigate them. We also discuss electrically charged drugs that are currently being evaluated in clinical trials and identify areas of further development that have the potential to usher in new treatments.

19.
J Vis Exp ; (162)2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32831304

RESUMO

Several negatively charged tissues in the body, like cartilage, present a barrier to the targeted drug delivery due to their high density of negatively charged aggrecans and, therefore, require improved targeting methods to increase their therapeutic response. Because cartilage has a high negative fixed charge density, drugs can be modified with positively charged drug carriers to take advantage of electrostatic interactions, allowing for enhanced intra-cartilage drug transport. Studying the transport of drug carriers is, therefore, crucial towards predicting the efficacy of drugs in inducing a biological response. We show the design of three experiments which can quantify the equilibrium uptake, depth of penetration and non-equilibrium diffusion rate of cationic peptide carriers in cartilage explants. Equilibrium uptake experiments provide a measure of the solute concentration within the cartilage compared to its surrounding bath, which is useful for predicting the potential of a drug carrier in enhancing therapeutic concentration of drugs in cartilage. Depth of penetration studies using confocal microscopy allow for the visual representation of 1D solute diffusion from the superficial to deep zone of cartilage, which is important for assessing whether solutes reach their matrix and cellular target sites. Non-equilibrium diffusion rate studies using a custom-designed transport chamber enables the measurement of the strength of binding interactions with the tissue matrix by characterizing the diffusion rates of fluorescently labeled solutes across the tissue; this is beneficial for designing carriers of optimal binding strength with cartilage. Together, the results obtained from the three transport experiments provide a guideline for designing optimally charged drug carriers which take advantage of weak and reversible charge interactions for drug delivery applications. These experimental methods can also be applied to evaluate the transport of drugs and drug-drug carrier conjugates. Further, these methods can be adapted for the use in targeting other negatively charged tissues such as meniscus, cornea and the vitreous humor.


Assuntos
Cartilagem/metabolismo , Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Peptídeos/farmacocinética , Animais , Cartilagem/efeitos dos fármacos , Cátions/química , Difusão , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Peptídeos/administração & dosagem , Peptídeos/química , Eletricidade Estática
20.
Sci Rep ; 10(1): 12017, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32694557

RESUMO

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.


Assuntos
Avidina/administração & dosagem , Dextranos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Disco Intervertebral/efeitos dos fármacos , Nanoestruturas/administração & dosagem , Animais , Avidina/farmacologia , Bovinos , Dextranos/farmacologia , Glicosaminoglicanos , Meia-Vida , Injeções , Degeneração do Disco Intervertebral/tratamento farmacológico , Dor Lombar/tratamento farmacológico , Núcleo Pulposo/efeitos dos fármacos , Núcleo Pulposo/metabolismo , Eletricidade Estática
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...