Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.904
Filter
1.
Int J Nanomedicine ; 19: 7307-7321, 2024.
Article in English | MEDLINE | ID: mdl-39050879

ABSTRACT

Background: Challenges such as poor drug selectivity, non-target reactivity, and the development of drug resistance continue to pose significant obstacles in the clinical application of cancer therapeutic drugs. To overcome the limitations of drug resistance in chemotherapy, a viable treatment strategy involves designing multifunctional nano-platforms that exploit the unique physicochemical properties of tumor microenvironment (TME). Methods: Herein, layer-by-layer nanoparticles with polyporous CuS as delivery vehicles, loaded with a sonosensitizer (tetra-(4-aminophenyl) porphyrin, TAPP) and sequentially functionalized with pH-responsive CaCO3, targeting group hyaluronic acid (HA) were designed and synthesized for synergistic treatment involving chemodynamic therapy (CDT), sonodynamic therapy (SDT), photothermal therapy (PTT), and calcium overload. Upon cleavage in an acidic environment, CaCO3 nanoparticles released TAPP and Ca2+, with TAPP generating 1O2 under ultrasound trigger. Exposed CuS produced highly cytotoxic ·OH in response to H2O2 and also exhibited a strong PTT effect. Results: CuS@TAPP-CaCO3/HA (CTCH) delivered an enhanced ability to release more Ca2+ under acidic conditions with a pH value of 6.5, which in situ causes damage to HeLa mitochondria. In vitro and in vivo experiments both demonstrated that mitochondrial dysfunction greatly amplified the damage caused by reactive oxygen species (ROS) to tumor, which strongly confirms the synergistic effect between calcium overload and reactive oxygen therapy. Conclusion: Collectively, the development of CTCH presents a novel therapeutic strategy for tumor treatment by effectively responding to the acidic TME, thus holding significant clinical implications.


Subject(s)
Calcium Carbonate , Calcium , Nanoparticles , Tumor Microenvironment , Humans , Animals , Nanoparticles/chemistry , Calcium/chemistry , Calcium Carbonate/chemistry , Calcium Carbonate/pharmacology , Tumor Microenvironment/drug effects , HeLa Cells , Reactive Oxygen Species/metabolism , Mice , Hyaluronic Acid/chemistry , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/pharmacokinetics , Porphyrins/administration & dosage , Photothermal Therapy/methods , Hydrogen-Ion Concentration , Ultrasonic Therapy/methods , Neoplasms/therapy , Neoplasms/drug therapy , Mitochondria/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C , Mice, Nude , Layer-by-Layer Nanoparticles
2.
Skin Res Technol ; 30(7): e13838, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39036993

ABSTRACT

BACKGROUND: Understanding the differences in soft tissue filler rheology and how these properties can impact clinical results is a fundamental concepts for any injector. This study aimed to assess the tissue integration characteristics of hyaluronic acid (HA) fillers manufactured with different technologies (Non-Animal Stabilized HA [HA-N] or Optimal Balance Technology [HA-O]) using ultra-high-frequency ultrasound. METHODS: Twelve female participants with mild-to-moderate midface volume loss and temporal hollowing were enrolled and treated with HA-N and/or HA-O. Participants were seen at five visits (screening/baseline [treatment], and Weeks 1 [optional touch-up], 4, 6, and 8 [follow-up visits]). Ultrasound was used to evaluate the degree of product integration. RESULTS: On ultrasound, HA-N presented with distinct borders, minimal tissue integration, and a capacity to displace tissues. Conversely, HA-O tended to spread horizontally within the same tissue plane and integrated within tissues. The volumizing capacity of the HA-O fillers was dependent on particle size. CONCLUSION: HA-N is suited for deep injections in areas such as the upper lateral cheek and under the muscle of the temporal region when a lifting effect is desired; HA-O is best suited for subcutaneous injections, in areas of dynamic movement or for patients with thin skin; and can be injected subcutaneously or supraperiosteally when a volumizing effect is desired.


Subject(s)
Cosmetic Techniques , Dermal Fillers , Hyaluronic Acid , Rheology , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/chemistry , Humans , Female , Dermal Fillers/administration & dosage , Dermal Fillers/chemistry , Middle Aged , Adult , Esthetics , Ultrasonography/methods , Aged , Skin Aging/drug effects , Face/diagnostic imaging , Particle Size
3.
ACS Appl Mater Interfaces ; 16(28): 36142-36156, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38968001

ABSTRACT

There is an urgent need to develop phototherapeutic agents with imaging capabilities to assess the treatment process and efficacy in real-time during cancer phototherapy for precision cancer therapy. The safe near-infrared (NIR) fluorescent dyes have garnered significant attention and are desirable for theranostics agents. However, until now, achieving excellent photostability and fluorescence (FL) imaging capability in aggregation-caused quenching (ACQ) dyes remains a big challenge. Here, for the only FDA-approved NIR dye, indocyanine green (ICG), we developed a dual-ferrocene (Fc) chimeric nanonetwork ICG@HFFC based on the rigid-flexible strategy through one-step self-assembly, which uses rigid Fc-modified hyaluronic acid (HA) copolymer (HA-Fc) and flexible octadecylamine (ODA) bonded Fc (Fc-C18) as the delivery system. HA-Fc reserved the ability of HA to target the CD44 receptor of the tumor cell surface, and the dual-Fc region provided a rigid space for securely binding ICG through metal-ligand interaction and π-π conjugation, ensuring excellent photostability. Additionally, the alkyl chain provided flexible confinement for the remaining ICG through hydrophobic forces, preserving its FL. Thereby, a balance is achieved between outstanding photostability and FL imaging capability. In vitro studies showed improved photobleaching resistance, enhanced FL stability, and increased singlet oxygen (1O2) production efficiency in ICG@HFFC. Further in vivo results display that ICG@HFFC had good tumor tracing ability and significant tumor inhibition which also exhibited good biocompatibility.. Therefore, ICG@HFFC provides an encouraging strategy to realize simultaneous enhanced tumor tracing and photothermal/photodynamic therapy (PTT/PDT) and offers a novel approach to address the limitations of ACQ dyes.


Subject(s)
Ferrous Compounds , Hyaluronic Acid , Indocyanine Green , Metallocenes , Photochemotherapy , Ferrous Compounds/chemistry , Humans , Metallocenes/chemistry , Animals , Mice , Indocyanine Green/chemistry , Indocyanine Green/therapeutic use , Indocyanine Green/pharmacology , Hyaluronic Acid/chemistry , Photothermal Therapy , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Mice, Nude , Cell Line, Tumor , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Nanoparticles/chemistry , Nanoparticles/therapeutic use
4.
ACS Appl Mater Interfaces ; 16(28): 35949-35963, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38970482

ABSTRACT

Chemotherapy-induced oral mucositis (CIOM) is a prevalent complication of chemotherapy and significantly affects the treatment process. However, effective treatment for CIOM is lacking due to the unique environment of the oral cavity and the single effect of current drug delivery systems. In this present study, we propose an innovative approach by combining a methacrylate-modified human recombinant collagen III (rhCol3MA) hydrogel system with hyaluronic acid-epigallocatechin gallate (HA-E) and dopamine-modified methacrylate-alginate (AlgDA-MA). HA-E is used as an antioxidant and anti-inflammatory agent and synergizes with AlgDA-MA to improve the wet adhesion of hydrogel. The results of rhCol3MA/HA-E/AlgDA-MA (Col/HA-E/Alg) hydrogel demonstrate suitable physicochemical properties, excellent wet adhesive capacity, and biocompatibility. Notably, the hydrogel could promote macrophage polarization from M1 to M2 and redress human oral keratinocyte (HOK) inflammation by inhibiting NF-κB activation. Wound healing evaluations in vivo demonstrate that the Col/HA-E/Alg hydrogel exhibits a pro-repair effect by mitigating inflammatory imbalances, fostering early angiogenesis, and facilitating collagen repair. In summary, the Col/HA-E/Alg hydrogel could serve as a promising multifunctional dressing for the treatment of CIOM.


Subject(s)
Alginates , Anti-Inflammatory Agents , Hyaluronic Acid , Hydrogels , Stomatitis , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Stomatitis/drug therapy , Stomatitis/chemically induced , Stomatitis/pathology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Alginates/chemistry , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Catechin/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/therapeutic use , Mice , Wound Healing/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Methacrylates/chemistry , Dopamine/chemistry , Dopamine/pharmacology , Keratinocytes/drug effects
5.
J Biomed Mater Res B Appl Biomater ; 112(8): e35453, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39021285

ABSTRACT

Rheumatoid arthritis (RA) requires therapeutic approaches that alleviate symptoms and inhibit the progression of joint damage. Glucocorticoids (GCs) have been a cornerstone of RA treatment, yet their use is often limited by side effects. Recent advancements suggest that liposome-based delivery systems can improve GC biodistribution, minimizing toxicity. This study introduces an innovative tool for RA treatment using prednisone-encapsulated nonphospholipid liposomes (NPLs) in combination with a hyaluronic acid (HA) hydrogel. Our methodology involved incorporating prednisone (PR) with palmitic acid and cholesterol to formulate stable NPLs using a thin-film hydration technique. The synthesized PR-NPLs, characterized by a mean size of 150 nm, demonstrated uniform distribution and higher drug encapsulation in comparison with conventional phospholipid liposomes. In vitro assays revealed that PR-NPL markedly reduced inflammatory responses in macrophages. Additionally, we successfully incorporated PR-NPL into an HA hydrogel, employing a photoinitiated cross-linking process. This novel composite offered modulable PR release, governed by the degree of hydrogel cross-linking. The developed system presents a promising advancement in RA management, especially suited for intraarticular injections. It potentially enables targeted, controlled drug release with a reduced risk of side effects, signifying a significant improvement over existing RA therapies.


Subject(s)
Arthritis, Rheumatoid , Hyaluronic Acid , Hydrogels , Liposomes , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Liposomes/chemistry , Arthritis, Rheumatoid/drug therapy , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Animals , Prednisolone/chemistry , Prednisolone/pharmacology , Prednisolone/pharmacokinetics , Humans , RAW 264.7 Cells
6.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000293

ABSTRACT

Cell mechanics are a biophysical indicator of cell state, such as cancer metastasis, leukocyte activation, and cell cycle progression. Atomic force microscopy (AFM) is a widely used technique to measure cell mechanics, where the Young modulus of a cell is usually derived from the Hertz contact model. However, the Hertz model assumes that the cell is an elastic, isotropic, and homogeneous material and that the indentation is small compared to the cell size. These assumptions neglect the effects of the cytoskeleton, cell size and shape, and cell environment on cell deformation. In this study, we investigated the influence of cell size on the estimated Young's modulus using liposomes as cell models. Liposomes were prepared with different sizes and filled with phosphate buffered saline (PBS) or hyaluronic acid (HA) to mimic the cytoplasm. AFM was used to obtain the force indentation curves and fit them to the Hertz model. We found that the larger the liposome, the lower the estimated Young's modulus for both PBS-filled and HA-filled liposomes. This suggests that the Young modulus obtained from the Hertz model is not only a property of the cell material but also depends on the cell dimensions. Therefore, when comparing or interpreting cell mechanics using the Hertz model, it is essential to account for cell size.


Subject(s)
Elastic Modulus , Liposomes , Microscopy, Atomic Force , Microscopy, Atomic Force/methods , Liposomes/chemistry , Cell Size , Models, Biological , Hyaluronic Acid/chemistry , Biomechanical Phenomena , Humans
7.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38999964

ABSTRACT

Keeping wounds clean in small animals is a big challenge, which is why they often become infected, creating a risk of transmission to animal owners. Therefore, it is crucial to search for new biocompatible materials that have the potential to be used in smart wound dressings with both wound healing and bacteriostatic properties to prevent infection. In our previous work, we obtained innovative hyaluronate matrix-based bionanocomposites containing nanosilver and nanosilver/graphene oxide (Hyal/Ag and Hyal/Ag/GO). This study aimed to thoroughly examine the bacteriostatic properties of foils containing the previously developed bionanocomposites. The bacteriostatic activity was assessed in vitro on 88 Gram-positive (n = 51) and Gram-negative (n = 37) bacteria isolated from wounds of small animals and whose antimicrobial resistance patterns and resistance mechanisms were examined in an earlier study. Here, 69.32% of bacterial growth was inhibited by Hyal/Ag and 81.82% by Hyal/Ag/GO. The bionanocomposites appeared more effective against Gram-negative bacteria (growth inhibition of 75.68% and 89.19% by Hyal/Ag and Hyal/Ag/Go, respectively). The effectiveness of Hyal/Ag/GO against Gram-positive bacteria was also high (inhibition of 80.39% of strains), while Hyal/Ag inhibited the growth of 64.71% of Gram-positive bacteria. The effectiveness of Hyal/Ag and Hyal/Ag/Go varied depending on bacterial genus and species. Proteus (Gram-negative) and Enterococcus (Gram-positive) appeared to be the least susceptible to the bionanocomposites. Hyal/Ag most effectively inhibited the growth of non-pathogenic Gram-positive Sporosarcina luteola and Gram-negative Acinetobacter. Hyal/Ag/GO was most effective against Gram-positive Streptococcus and Gram-negative Moraxella osloensis. The Hyal/Ag/GO bionanocomposites proved to be very promising new antibacterial, biocompatible materials that could be used in the production of bioactive wound dressings.


Subject(s)
Anti-Bacterial Agents , Graphite , Hyaluronic Acid , Metal Nanoparticles , Microbial Sensitivity Tests , Nanocomposites , Silver , Graphite/chemistry , Graphite/pharmacology , Nanocomposites/chemistry , Metal Nanoparticles/chemistry , Silver/chemistry , Silver/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Animals , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Wound Healing/drug effects , Bacteria/drug effects , Bacteria/growth & development
8.
Biosens Bioelectron ; 262: 116539, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38950517

ABSTRACT

Prostaglandin E2 (PGE2), an eicosane, regulates the physiological activity of inflammatory cells and represents a potential therapeutic target for facilitating tissue repair in vivo. In our work, an electrochemical immunosensor employing Ketjen black-Au nanoparticles (KB-Au) and poly tannic acid nanospheres conjugated with anti-PGE2 polyclonal antibody (PTAN-Ab) was designed to ultra-sensitively analyze PGE2 levels secreted by living cells and tissues. Antibody assembly strategies were explored to achieve signal amplification. Moreover, we studied the therapy effects of docosahexaenoic acid (DHA), arachidonic acid (AA), hyaluronic acid (HA), and small molecule 15-hydroxyprostaglandin dehydrogenase inhibitor (SW033291) on inflammation and evaluated the protective functions of HA and SW033291 in a murine model subjected to colitis induced by dextran sulfate sodium (DSS) using the developed sensor. The sensor exhibited a linear range of 10-5-106 fg/mL and a detection limit (LOD) of 10-5 fg/mL. Fetal bovine serum (FBS) samples were used to achieve high recovery of target analytes. This study not only presents an effective strategy for ultra-sensitively monitoring PGE2 but also provides valuable insights into assessing the degree of inflammation and the therapeutic effect of related drugs. Research on human health monitoring and regenerative medicine could greatly benefit from the findings.


Subject(s)
Biosensing Techniques , Dinoprostone , Electrochemical Techniques , Inflammation , Animals , Mice , Dinoprostone/analysis , Electrochemical Techniques/methods , Inflammation/drug therapy , Humans , Gold/chemistry , Metal Nanoparticles/chemistry , Limit of Detection , Colitis/drug therapy , Hyaluronic Acid/chemistry , Tannins/chemistry , Immunoassay/methods
9.
Microb Cell Fact ; 23(1): 200, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026213

ABSTRACT

Hyaluronidase (hyase) is an endoglycosidase enzyme that degrades hyaluronic acid (HA) and is mostly known to be found in the extracellular matrix of connective tissues. In the current study, eleven bacteria isolates and one actinomycete were isolated from a roaster comb and screened for hyase production. Seven isolates were positive for hyase, and the most potent isolate was selected based on the diameter of the transparent zone. Based on the morphological, physiological, and 16 S rRNA characteristics, the most potent isolate was identified as Brucella intermedia MEFS with accession number OR794010. The environmental conditions supporting the maximum production of hyase were optimized to be incubation at 30 ºC for 48 h and pH 7, which caused a 1.17-fold increase in hyase production with an activity of 84 U/mL. Hyase was purified using a standard protocol, including precipitation with ammonium sulphate, DEAE as ion exchange chromatography, and size exclusion chromatography using Sephacryle S100, with a specific activity of 9.3-fold compared with the crude enzyme. The results revealed that the molecular weight of hyase was 65 KDa, and the optimum conditions for hyase activity were at pH 7.0 and 37 °C for 30 min. The purified hyase showed potent anticancer activities against colon, lung, skin, and breast cancer cell lines with low toxicity against normal somatic cells. The cell viability of hyase-treated cancer cells was found to be in a dose dependent manner. Hyase also controlled the growth factor-induced cell cycle progression of breast cancer cells and caused relative changes in angiogenesis-related genes as well as suppressed many pro-inflammatory proteins in MDA cells compared with 5-fluorouracil, indicating the significant role of hyase as an anticancer agent. In addition, hyase recorded the highest DPPH scavenging activity of 65.49% and total antioxidant activity of 71.84% at a concentration of 200 µg/mL.


Subject(s)
Antineoplastic Agents , Antioxidants , Hyaluronoglucosaminidase , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/antagonists & inhibitors , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antioxidants/pharmacology , Antioxidants/metabolism , Antioxidants/chemistry , Cell Line, Tumor , Hydrogen-Ion Concentration , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/chemistry
10.
Nat Commun ; 15(1): 5902, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003266

ABSTRACT

Analogous of pixels to two-dimensional pictures, voxels-in the form of either small cubes or spheres-are the basic building blocks of three-dimensional objects. However, precise manipulation of viscoelastic bio-ink voxels in three-dimensional space represents a grand challenge in both soft matter science and biomanufacturing. Here, we present a voxelated bioprinting technology that enables the digital assembly of interpenetrating double-network hydrogel droplets made of polyacrylamide/alginate-based or hyaluronic acid/alginate-based polymers. The hydrogels are crosslinked via additive-free and biofriendly click reaction between a pair of stoichiometrically matched polymers carrying norbornene and tetrazine groups, respectively. We develop theoretical frameworks to describe the crosslinking kinetics and stiffness of the hydrogels, and construct a diagram-of-state to delineate their mechanical properties. Multi-channel print nozzles are developed to allow on-demand mixing of highly viscoelastic bio-inks without significantly impairing cell viability. Further, we showcase the distinctive capability of voxelated bioprinting by creating highly complex three-dimensional structures such as a hollow sphere composed of interconnected yet distinguishable hydrogel particles. Finally, we validate the cytocompatibility and in vivo stability of the printed double-network scaffolds through cell encapsulation and animal transplantation.


Subject(s)
Acrylic Resins , Alginates , Bioprinting , Hyaluronic Acid , Hydrogels , Bioprinting/methods , Hydrogels/chemistry , Alginates/chemistry , Animals , Hyaluronic Acid/chemistry , Acrylic Resins/chemistry , Mice , Ink , Printing, Three-Dimensional , Humans , Tissue Engineering/methods , Cell Survival , Biocompatible Materials/chemistry
11.
J Nanobiotechnology ; 22(1): 383, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951875

ABSTRACT

The characteristic features of the rheumatoid arthritis (RA) microenvironment are synovial inflammation and hyperplasia. Therefore, there is a growing interest in developing a suitable therapeutic strategy for RA that targets the synovial macrophages and fibroblast-like synoviocytes (FLSs). In this study, we used graphene oxide quantum dots (GOQDs) for loading anti-arthritic sinomenine hydrochloride (SIN). By combining with hyaluronic acid (HA)-inserted hybrid membrane (RFM), we successfully constructed a new nanodrug system named HA@RFM@GP@SIN NPs for target therapy of inflammatory articular lesions. Mechanistic studies showed that this nanomedicine system was effective against RA by facilitating the transition of M1 to M2 macrophages and inhibiting the abnormal proliferation of FLSs in vitro. In vivo therapeutic potential investigation demonstrated its effects on macrophage polarization and synovial hyperplasia, ultimately preventing cartilage destruction and bone erosion in the preclinical models of adjuvant-induced arthritis and collagen-induced arthritis in rats. Metabolomics indicated that the anti-arthritic effects of HA@RFM@GP@SIN NPs were mainly associated with the regulation of steroid hormone biosynthesis, ovarian steroidogenesis, tryptophan metabolism, and tyrosine metabolism. More notably, transcriptomic analyses revealed that HA@RFM@GP@SIN NPs suppressed the cell cycle pathway while inducing the cell apoptosis pathway. Furthermore, protein validation revealed that HA@RFM@GP@SIN NPs disrupted the excessive growth of RAFLS by interfering with the PI3K/Akt/SGK/FoxO signaling cascade, resulting in a decline in cyclin B1 expression and the arrest of the G2 phase. Additionally, considering the favorable biocompatibility and biosafety, these multifunctional nanoparticles offer a promising therapeutic approach for patients with RA.


Subject(s)
Arthritis, Rheumatoid , Cell Proliferation , Graphite , Macrophages , Morphinans , Quantum Dots , Synoviocytes , Morphinans/pharmacology , Morphinans/chemistry , Animals , Quantum Dots/chemistry , Quantum Dots/therapeutic use , Arthritis, Rheumatoid/drug therapy , Synoviocytes/drug effects , Synoviocytes/metabolism , Graphite/chemistry , Graphite/pharmacology , Cell Proliferation/drug effects , Rats , Macrophages/drug effects , Macrophages/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Rats, Sprague-Dawley , Mice , Humans , RAW 264.7 Cells , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology
12.
Mikrochim Acta ; 191(8): 447, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38963544

ABSTRACT

An intelligent nanodrug delivery system (Cu/ZIF-8@GOx-DOX@HA, hereafter CZGDH) consisting of Cu-doped zeolite imidazolate framework-8 (Cu/ZIF-8, hereafter CZ), glucose oxidase (GOx), doxorubicin (DOX), and hyaluronic acid (HA) was established for targeted drug delivery and synergistic therapy of tumors. The CZGDH specifically entered tumor cells through the targeting effect of HA and exhibited acidity-triggered biodegradation for subsequent release of GOx, DOX, and Cu2+ in the tumor microenvironment (TME). The GOx oxidized the glucose (Glu) in tumor cells to produce H2O2 and gluconic acid for starvation therapy (ST). The DOX entered the intratumoral cell nucleus for chemotherapy (CT). The released Cu2+ consumed the overexpressed glutathione (GSH) in tumor cells to produce Cu+. The generated Cu+ and H2O2 triggered the Fenton-like reaction to generate toxic hydroxyl radicals (·OH), which disrupted the redox balance of tumor cells and effectively killed tumor cells for chemodynamic therapy (CDT). Therefore, synergistic multimodal tumor treatment via TME-activated cascade reaction was achieved. The nanodrug delivery system has a high drug loading rate (48.3 wt%), and the three-mode synergistic therapy has a strong killing effect on tumor cells (67.45%).


Subject(s)
Copper , Doxorubicin , Glucose Oxidase , Hyaluronic Acid , Metal-Organic Frameworks , Tumor Microenvironment , Zeolites , Copper/chemistry , Doxorubicin/pharmacology , Doxorubicin/chemistry , Tumor Microenvironment/drug effects , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Humans , Zeolites/chemistry , Animals , Metal-Organic Frameworks/chemistry , Hyaluronic Acid/chemistry , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Cell Line, Tumor , Mice , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Neoplasms/drug therapy , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Imidazoles
13.
Sci Adv ; 10(27): eado9120, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959311

ABSTRACT

A bioinspired hydrogel composed of hyaluronic acid-graft-dopamine (HADA) and a designer peptide HGF-(RADA)4-DGDRGDS (HRR) was presented to enhance tissue integration following spinal cord injury (SCI). The HADA/HRR hydrogel manipulated the infiltration of PDGFRß+ cells in a parallel pattern, transforming dense scars into an aligned fibrous substrate that guided axonal regrowth. Further incorporation of NT3 and curcumin promoted axonal regrowth and survival of interneurons at lesion borders, which served as relays for establishing heterogeneous axon connections in a target-specific manner. Notable improvements in motor, sensory, and bladder functions resulted in rats with complete spinal cord transection. The HADA/HRR + NT3/Cur hydrogel promoted V2a neuron accumulation in ventral spinal cord, facilitating the recovery of locomotor function. Meanwhile, the establishment of heterogeneous neural connections across the hemisected lesion of canines was documented in a target-specific manner via neuronal relays, significantly improving motor functions. Therefore, biomaterials can inspire beneficial biological activities for SCI repair.


Subject(s)
Extracellular Matrix , Hydrogels , Spinal Cord Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , Hydrogels/chemistry , Rats , Extracellular Matrix/metabolism , Neurons/metabolism , Neurons/drug effects , Dogs , Axons/metabolism , Axons/drug effects , Nerve Regeneration/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Recovery of Function/drug effects , Dopamine/metabolism , Female , Disease Models, Animal , Rats, Sprague-Dawley , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Spinal Cord/metabolism
14.
J Nanobiotechnology ; 22(1): 432, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39034393

ABSTRACT

Rheumatoid arthritis (RA) involves chronic joint inflammation. Combining acupuncture and medication for RA treatment faces challenges like spatiotemporal variability, limited drug loading in acupuncture needles, and premature or untargeted drug release. Here, we designed a new type of tubular acupuncture needles, with an etched hollow honeycomb-like structure to enable the high loading of therapeutics, integrating the traditional acupuncture and drug repository into an all-in-one therapeutic platform. In these proof-of-concept experiments, we fabricated injectable hollow honeycomb electroacupuncture needles (HC-EA) loaded with melittin hydrogel (MLT-Gel), enabling the combination treatment of acupuncture stimulation and melittin therapy in a spatiotemporally synchronous manner. Since the RA microenvironment is mildly acidic, the acid-responsive chitosan (CS)/sodium beta-glycerophosphate (ß-GP)/ hyaluronic acid (HA) composited hydrogel (CS/GP/HA) was utilized to perform acupuncture stimulation and achieve the targeted release of injected therapeutics into the specific lesion site. Testing our therapeutic platform involved a mouse model of RA and bioinformatics analysis. MLT-Gel@HC-EA treatment restored Th17/Treg-mediated immunity balance, reduced inflammatory factor release (TNF-α, IL-6, IL-1ß), and alleviated inflammation at the lesion site. This novel combination of modified acupuncture needle and medication, specifically melittin hydrogel, holds promise as a therapeutic strategy for RA treatment.


Subject(s)
Acupuncture Therapy , Arthritis, Rheumatoid , Hydrogels , Melitten , Needles , Animals , Melitten/pharmacology , Melitten/chemistry , Mice , Arthritis, Rheumatoid/therapy , Arthritis, Rheumatoid/drug therapy , Hydrogels/chemistry , Acupuncture Therapy/methods , Chitosan/chemistry , Hyaluronic Acid/chemistry , Male , Disease Models, Animal , Mice, Inbred C57BL
15.
J Mater Chem B ; 12(29): 7122-7134, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38946474

ABSTRACT

Micro- and nanoencapsulation of cells has been studied as a strategy to protect cells from environmental stress and promote survival during delivery. Hydrogels used in encapsulation can be modified to influence cell behaviors and direct assembly in their surroundings. Here, we report a system that conformally encapsulated stem cells using hyaluronic acid (HA). We successfully modified HA with lipid, thiol, and maleimide pendant groups to facilitate a hydrogel system in which HA was deposited onto cell plasma membranes and subsequently crosslinked through thiol-maleimide click chemistry. We demonstrated conformal encapsulation of both neural stem cells (NSCs) and mesenchymal stromal cells (MSCs), with viability of both cell types greater than 90% after encapsulation. Additional material could be added to the conformal hydrogel through alternating addition of thiol-modified and maleimide-modified HA in a layering process. After encapsulation, we tracked egress and viability of the cells over days and observed differential responses of cell types to conformal hydrogels both according to cell type and the amount of material deposited on the cell surfaces. Through the design of the conformal hydrogels, we showed that multicellular assembly could be created in suspension and that encapsulated cells could be immobilized on surfaces. In conjunction with photolithography, conformal hydrogels enabled rapid assembly of encapsulated cells on hydrogel substrates with resolution at the scale of 100 µm.


Subject(s)
Cell Survival , Hyaluronic Acid , Hydrogels , Mesenchymal Stem Cells , Neural Stem Cells , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Cell Survival/drug effects , Cell Encapsulation/methods , Mice , Surface Properties , Cells, Cultured
16.
Expert Opin Drug Deliv ; 21(6): 965-974, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38962819

ABSTRACT

OBJECTIVE: Dissolving microneedles (DMNs) have shown great potential for transdermal drug delivery due to their excellent skin-penetrating ability and combination with nanocarriers (NCs) can realize targeted drug delivery. The objective of this study was to investigate the impact of microneedle dissolving rate on the in vivo fate of NC-loaded DMNs, which would facilitate the clinical translation of such systems. METHODS: Solid lipid nanoparticles (SLNs) were selected as the model NC for loading in DMNs, which were labeled by P4 probes with aggregation-quenching properties. Sodium hyaluronate acid (HA) and chitosan (CS), with different aqueous dissolving rates, were chosen as model tip materials. The effects of needle dissolving rate on the in vivo fate of NC-loaded DMNs was investigated by tracking the distribution of fluorescence signals after transdermal exposure. RESULTS: P4 SLNs achieved a deeper diffusion depth of 180 µm in DMN-HA with a faster dissolution rate, while the diffusion depth in DMN-CS with a slower dissolution rate was lower (140 µm). The in vivo experiments demonstrated that P4 SLNs had a T1/2 value of 12.14 h in DMN-HA, whilst a longer retention time was found in DMN-CS, with a T1/2 of 13.12 h. CONCLUSIONS: This study confirmed that the in vivo diffusion rate of NC-loaded DMNs was determined by the dissolving rate of DMNs materials and provided valuable guidance for the design and development of NC-loaded DMNs in the future.


Subject(s)
Administration, Cutaneous , Chitosan , Drug Delivery Systems , Hyaluronic Acid , Nanoparticles , Needles , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/administration & dosage , Chitosan/chemistry , Drug Carriers/chemistry , Solubility , Lipids/chemistry , Microinjections , Skin Absorption , Skin/metabolism , Male , Rats, Sprague-Dawley , Rats , Liposomes
17.
Nat Commun ; 15(1): 5689, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38971796

ABSTRACT

Leukemia is a kind of hematological malignancy originating from bone marrow, which provides essential signals for initiation, progression, and recurrence of leukemia. However, how to specifically deliver drugs to the bone marrow remains elusive. Here, we develop biomimetic vesicles by infusing hematopoietic stem and progenitor cell (HSPC) membrane with liposomes (HSPC liposomes), which migrate to the bone marrow of leukemic mice via hyaluronic acid-CD44 axis. Moreover, the biomimetic vesicles exhibit superior binding affinity to leukemia cells through intercellular cell adhesion molecule-1 (ICAM-1)/integrin ß2 (ITGB2) interaction. Further experiments validate that the vesicles carrying chemotherapy drug cytarabine (Ara-C@HSPC-Lipo) markedly inhibit proliferation, induce apoptosis and differentiation of leukemia cells, and decrease number of leukemia stem cells. Mechanically, RNA-seq reveals that Ara-C@HSPC-Lipo treatment induces apoptosis and differentiation and inhibits the oncogenic pathways. Finally, we verify that HSPC liposomes are safe in mice. This study provides a method for targeting bone marrow and treating leukemia.


Subject(s)
Apoptosis , Bone Marrow , Cytarabine , Drug Delivery Systems , Hematopoietic Stem Cells , Leukemia , Liposomes , Animals , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Mice , Cytarabine/pharmacology , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/metabolism , Apoptosis/drug effects , Leukemia/drug therapy , Leukemia/pathology , Humans , Cell Differentiation/drug effects , Cell Membrane/metabolism , Cell Membrane/drug effects , Cell Line, Tumor , CD18 Antigens/metabolism , Cell Proliferation/drug effects , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism
18.
Sci Adv ; 10(30): eado8222, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058786

ABSTRACT

The artificial mucus layer, such as hydrogels, used to repair the damaged intestinal barrier, is a promising treatment for inflammatory bowel disease (IBD). However, the currently reported hydrogel-based artificial barriers are administered via rectal injection, causing unnecessary discomfort to patients. Herein, we report an oral hydrogel precursor solution based on thiol-modified hyaluronic acid (HASH). Owing to the reactive oxygen species (ROS)-responsive gelling behavior, our precursor solution formed an artificial mucus coating over the inflamed regions of the intestines, blocking microbial invasion and reducing abnormally activated immune responses. Notably, HASH also modulated the gut microbiota, including increasing the diversity and enhancing the abundance of short-chain fatty acid-associated bacteria, which play a key role in gut homeostasis. We believe that the ROS-responsive artificial mucus layer is a promising strategy for the oral treatment of IBD.


Subject(s)
Hyaluronic Acid , Inflammatory Bowel Diseases , Mucus , Reactive Oxygen Species , Reactive Oxygen Species/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Mucus/metabolism , Animals , Hyaluronic Acid/chemistry , Administration, Oral , Mice , Gastrointestinal Microbiome/drug effects , Hydrogels/chemistry , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Disease Models, Animal
19.
Biomed Pharmacother ; 177: 117051, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959608

ABSTRACT

Due to the limitations of the current skin wound treatments, it is highly valuable to have a wound healing formulation that mimics the extracellular matrix (ECM) and mechanical properties of natural skin tissue. Here, a novel biomimetic hydrogel formulation has been developed based on a mixture of Agarose-Collagen Type I (AC) combined with skin ECM-related components: Dermatan sulfate (DS), Hyaluronic acid (HA), and Elastin (EL) for its application in skin tissue engineering (TE). Different formulations were designed by combining AC hydrogels with DS, HA, and EL. Cell viability, hemocompatibility, physicochemical, mechanical, and wound healing properties were investigated. Finally, a bilayered hydrogel loaded with fibroblasts and mesenchymal stromal cells was developed using the Ag-Col I-DS-HA-EL (ACDHE) formulation. The ACDHE hydrogel displayed the best in vitro results and acceptable physicochemical properties. Also, it behaved mechanically close to human native skin and exhibited good cytocompatibility. Environmental scanning electron microscopy (ESEM) analysis revealed a porous microstructure that allows the maintenance of cell growth and ECM-like structure production. These findings demonstrate the potential of the ACDHE hydrogel formulation for applications such as an injectable hydrogel or a bioink to create cell-laden structures for skin TE.


Subject(s)
Biomimetic Materials , Hydrogels , Tissue Engineering , Hydrogels/chemistry , Humans , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Tissue Engineering/methods , Cell Survival/drug effects , Mesenchymal Stem Cells/drug effects , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Wound Healing/drug effects , Collagen Type I/metabolism , Skin/drug effects , Skin/metabolism , Dermatan Sulfate/chemistry , Dermatan Sulfate/pharmacology , Fibroblasts/drug effects , Elastin/chemistry , Extracellular Matrix/metabolism , Biomimetics/methods , Sepharose/chemistry , Dermis/drug effects , Dermis/metabolism , Dermis/cytology , Animals
20.
Int J Nanomedicine ; 19: 7493-7508, 2024.
Article in English | MEDLINE | ID: mdl-39081895

ABSTRACT

Introduction: Lung cancer is the most common cancer worldwide, among which non-small cell lung cancer (NSCLC) accounts for about 80% of all lung cancers. Chemotherapy, a mainstay modality for NSCLC, has demonstrated restricted effectiveness due to the emergence of chemo-resistance and systemic side effects. Studies have indicated that combining chemotherapy with phototherapy, such as photodynamic therapy (PDT) and photothermal therapy (PTT), can enhance efficacy of therapy. In this work, an aminated mesoporous graphene oxide (rPGO)-protoporphyrin IX (PPIX)-hyaluronic acid (HA)@Osimertinib (AZD) nanodrug delivery system (rPPH@AZD) was successfully developed for combined chemotherapy/phototherapy for NSCLC. Methods: A pH/hyaluronidase-responsive nanodrug delivery system (rPPH@AZD) was prepared using mesoporous graphene oxide. Its morphology, elemental composition, surface functional groups, optical properties, in vitro drug release ability, photothermal properties, reactive oxygen species production, cellular uptake and cell viability were evaluated. In addition, the in vivo therapeutic effect, biocompatibility, and imaging capabilities of rPPH@AZD were verified by a tumor-bearing mouse model. Results: Aminated mesoporous graphene oxide (rPGO) plays a role as a drug delivery vehicle owing to its large specific surface area and ease of surface functionalization. rPGO exhibits excellent photothermal conversion properties under laser irradiation, while PPIX acts as a photosensitizer to generate singlet oxygen. AZD acts as a small molecule targeted drug in chemotherapy. In essence, rPPH@AZD shows excellent photothermal and fluorescence imaging effects in tumor-bearing mice. More importantly, in vitro and in vivo results indicate that rPPH@AZD can achieve hyaluronidase/pH dual response as well as combined chemotherapy/PTT/PDT anti-NSCLC treatment. Conclusion: The newly prepared rPPH@AZD can serve as a promising pH/hyaluronidase-responsive nanodrug delivery system that integrates photothermal/fluorescence imaging and chemo/photo combined therapy for efficient therapy against NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Graphite , Hyaluronic Acid , Lung Neoplasms , Nanocomposites , Photochemotherapy , Graphite/chemistry , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Animals , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Humans , Mice , Nanocomposites/chemistry , Hyaluronic Acid/chemistry , Photochemotherapy/methods , Cell Line, Tumor , Protoporphyrins/chemistry , Protoporphyrins/pharmacokinetics , Cell Survival/drug effects , Drug Delivery Systems/methods , Combined Modality Therapy , Drug Liberation , Xenograft Model Antitumor Assays , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice, Nude , Porosity , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL