Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.171
Filter
1.
J Nanobiotechnology ; 22(1): 216, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698399

ABSTRACT

The enhanced permeability and retention (EPR) effect has become the guiding principle for nanomedicine against cancer for a long time. However, several biological barriers severely resist therapeutic agents' penetration and retention into the deep tumor tissues, resulting in poor EPR effect and high tumor mortality. Inspired by lava, we proposed a proteolytic enzyme therapy to improve the tumor distribution and penetration of nanomedicine. A trypsin-crosslinked hydrogel (Trypsin@PSA Gel) was developed to maintain trypsin's activity. The hydrogel postponed trypsin's self-degradation and sustained the release. Trypsin promoted the cellular uptake of nanoformulations in breast cancer cells, enhanced the penetration through endothelial cells, and degraded total and membrane proteins. Proteomic analysis reveals that trypsin affected ECM components and down-regulated multiple pathways associated with cancer progression. Intratumoral injection of Trypsin@PSA Gel significantly increased the distribution of liposomes in tumors and reduced tumor vasculature. Combination treatment with intravenous injection of gambogic acid-loaded liposomes and intratumoral injection of Trypsin@PSA Gel inhibited tumor growth. The current study provides one of the first investigations into the enhanced tumor distribution of liposomes induced by a novel proteolytic enzyme therapy.


Subject(s)
Hydrogels , Liposomes , Polyethylene Glycols , Trypsin , Xanthones , Liposomes/chemistry , Animals , Polyethylene Glycols/chemistry , Hydrogels/chemistry , Humans , Trypsin/metabolism , Trypsin/chemistry , Female , Mice , Cell Line, Tumor , Mice, Inbred BALB C , Breast Neoplasms/drug therapy , Proteolysis
2.
Sci Rep ; 14(1): 10073, 2024 05 02.
Article in English | MEDLINE | ID: mdl-38698123

ABSTRACT

Cutaneous leishmaniasis is the most prevalent form of leishmaniasis worldwide. Although various anti-leishmanial regimens have been considered, due to the lack of efficacy or occurrence of adverse reactions, design and development of novel topical delivery systems would be essential. This study aimed to prepare artemether (ART)-loaded niosomes and evaluate their anti-leishmanial effects against Leishmania major. ART-loaded niosomes were prepared through the thin-film hydration technique and characterized in terms of particle size, zeta potential, morphology, differential scanning calorimetry, drug loading, and drug release. Furthermore, anti-leishmanial effect of the preparation was assessed in vitro and in vivo. The prepared ART-loaded niosomes were spherical with an average diameter of about 100 and 300 nm with high encapsulation efficiencies of > 99%. The results of in vitro cytotoxicity revealed that ART-loaded niosomes had significantly higher anti-leishmanial activity, lower general toxicity, and higher selectivity index (SI). Half-maximal inhibitory concentration (IC50) values of ART, ART-loaded niosomes, and liposomal amphotericin B were 39.09, 15.12, and 20 µg/mL, respectively. Also, according to the in vivo study results, ART-loaded niosomes with an average size of 300 nm showed the highest anti-leishmanial effects in animal studies. ART-loaded niosomes would be promising topical drug delivery system for the management of cutaneous leishmaniasis.


Subject(s)
Artemether , Leishmania major , Leishmaniasis, Cutaneous , Liposomes , Liposomes/chemistry , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/parasitology , Artemether/chemistry , Leishmania major/drug effects , Animals , Mice , Particle Size , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/chemistry , Mice, Inbred BALB C , Drug Liberation , Humans
3.
Sci Rep ; 14(1): 10499, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714740

ABSTRACT

Improving the efficacy of chemotherapy remains a key challenge in cancer treatment, considering the low bioavailability, high cytotoxicity, and undesirable side effects of some clinical drugs. Targeted delivery and sustained release of therapeutic drugs to cancer cells can reduce the whole-body cytotoxicity of the agent and deliver a safe localized treatment to the patient. There is growing interest in herbal drugs, such as curcumin, which is highly noted as a promising anti-tumor drug, considering its wide range of bioactivities and therapeutic properties against various tumors. Conversely, the clinical efficacy of curcumin is limited because of poor oral bioavailability, low water solubility, instability in gastrointestinal fluids, and unsuitable pH stability. Drug-delivery colloid vehicles like liposomes and nanoparticles combined with microbubbles and ultrasound-mediated sustained release are currently being explored as effective delivery modes in such cases. This study aimed to synthesize and study the properties of curcumin liposomes (CLs) and optimize the high-frequency ultrasound release and uptake by a human breast cancer cell line (HCC 1954) through in vitro studies of culture viability and cytotoxicity. CLs were effectively prepared with particles sized at 81 ± 2 nm, demonstrating stability and controlled release of curcumin under ultrasound exposure. In vitro studies using HCC1954 cells, the combination of CLs, ultrasound, and Definity microbubbles significantly improved curcumin's anti-tumor effects, particularly under specific conditions: 15 s of continuous ultrasound at 0.12 W/cm2 power density with 0.6 × 107 microbubbles/mL. Furthermore, the study delved into curcumin liposomes' cytotoxic effects using an Annexin V/PI-based apoptosis assay. The treatment with CLs, particularly in conjunction with ultrasound and microbubbles, amplified cell apoptosis, mainly in the late apoptosis stage, which was attributed to heightened cellular uptake within cancer cells.


Subject(s)
Curcumin , Drug Delivery Systems , Liposomes , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/administration & dosage , Humans , Liposomes/chemistry , Cell Line, Tumor , Drug Delivery Systems/methods , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Microbubbles , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Ultrasonic Waves , Drug Liberation , Apoptosis/drug effects
4.
Molecules ; 29(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38731568

ABSTRACT

Cancer is one of the major causes of death, and its negative impact continues to rise globally. Chemotherapy, which is the most common therapy, has several limitations due to its tremendous side effects. Therefore, developing an alternate therapeutic agent with high biocompatibility is indeed needed. The anti-oxidative effects and bioactivities of several different crude extracts of marine algae have been evaluated both in vitro and in vivo. In the present study, we synthesized the aqueous extract (HA) from the marine algae Amphiroa anceps, and then, a liposome was formulated for that extract (NHA). The extracts were characterized using different photophysical tools like dynamic light scattering, UV-visible spectroscopy, FTIR, scanning electron microscopy, and GC-MS analysis. The SEM image revealed a size range of 112-185 nm for NHA and the GC-MS results showed the presence of octadecanoic acid and n-Hexadecanoic acid in the majority. The anticancer activity was studied using A549 cells, and the NHA inhibited the cancer cells dose-dependently, with the highest killing of 92% at 100 µg/mL. The in vivo studies in the zebrafish model showed that neither the HA nor NHA of Amphiroa anceps showed any teratogenic effect. The outcome of our study showed that NHA can be a potential drug candidate for inhibiting cancer with good biocompatibility up to a dose of 100 µg/mL.


Subject(s)
Antineoplastic Agents , Rhodophyta , Zebrafish , Rhodophyta/chemistry , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , A549 Cells , Neoplasms/drug therapy , Neoplasms/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Liposomes/chemistry , Gas Chromatography-Mass Spectrometry , Nanoparticles/chemistry , Cell Line, Tumor
5.
Int J Nanomedicine ; 19: 4121-4136, 2024.
Article in English | MEDLINE | ID: mdl-38736655

ABSTRACT

Purpose: This study aims to broaden the application of nano-contrast agents (NCAs) within the realm of the musculoskeletal system. It aims to introduce novel methods, strategies, and insights for the clinical management of ischemic muscle disorders, encompassing diagnosis, monitoring, evaluation, and therapeutic intervention. Methods: We developed a composite encapsulation technique employing O-carboxymethyl chitosan (OCMC) and liposome to encapsulate NCA-containing gold nanorods (GNRs) and perfluoropentane (PFP). This nanoscale contrast agent was thoroughly characterized for its basic physicochemical properties and performance. Its capabilities for in vivo and in vitro ultrasound imaging and photothermal imaging were authenticated, alongside a comprehensive biocompatibility assessment to ascertain its effects on microcirculatory perfusion in skeletal muscle using a murine model of hindlimb ischemia, and its potential to augment blood flow and facilitate recovery. Results: The engineered GNR@OCMC-liposome/PFP nanostructure exhibited an average size of 203.18±1.49 nm, characterized by size uniformity, regular morphology, and a good biocompatibility profile. In vitro assessments revealed NCA's potent photothermal response and its transformation into microbubbles (MBs) under near-infrared (NIR) irradiation, thereby enhancing ultrasonographic visibility. Animal studies demonstrated the nanostructure's efficacy in photothermal imaging at ischemic loci in mouse hindlimbs, where NIR irradiation induced rapid temperature increases and significantly increased blood circulation. Conclusion: The dual-modal ultrasound/photothermal NCA, encapsulating GNR and PFP within a composite shell-core architecture, was synthesized successfully. It demonstrated exceptional stability, biocompatibility, and phase transition efficiency. Importantly, it facilitates the encapsulation of PFP, enabling both enhanced ultrasound imaging and photothermal imaging following NIR light exposure. This advancement provides a critical step towards the integrated diagnosis and treatment of ischemic muscle diseases, signifying a pivotal development in nanomedicine for musculoskeletal therapeutics.


Subject(s)
Contrast Media , Gold , Ischemia , Muscle, Skeletal , Nanotubes , Ultrasonography , Animals , Gold/chemistry , Nanotubes/chemistry , Contrast Media/chemistry , Contrast Media/pharmacology , Mice , Ischemia/diagnostic imaging , Ischemia/therapy , Muscle, Skeletal/diagnostic imaging , Ultrasonography/methods , Hindlimb/blood supply , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Liposomes/chemistry , Chitosan/chemistry , Chitosan/pharmacology , Muscular Diseases/diagnostic imaging , Muscular Diseases/therapy , Photothermal Therapy/methods , Disease Models, Animal , Humans , Pentanes
6.
J Am Chem Soc ; 146(19): 12925-12932, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38691507

ABSTRACT

Technological breakthroughs in cryo-electron microscopy (cryo-EM) methods open new perspectives for highly detailed structural characterizations of extracellular vesicles (EVs) and synthetic liposome-protein assemblies. Structural characterizations of these vesicles in solution under a nearly native hydrated state are of great importance to decipher cell-to-cell communication and to improve EVs' application as markers in diagnosis and as drug carriers in disease therapy. However, difficulties in preparing holey carbon cryo-EM grids with low vesicle heterogeneities, at low concentration and with kinetic control of the chemical reactions or assembly processes, have limited cryo-EM use in the EV study. We report a straightforward membrane vesicle cryo-EM sample preparation method that assists in circumventing these limitations by using a free-standing DNA-affinity superlattice for covering holey carbon cryo-EM grids. Our approach uses DNA origami to self-assemble to a solution-stable and micrometer-sized ordered molecular template in which structure and functional properties can be rationally controlled. We engineered the template with cholesterol-binding sites to specifically trap membrane vesicles. The advantages of this DNA-cholesterol-affinity lattice (DCAL) include (1) local enrichment of artificial and biological vesicles at low concentration and (2) isolation of heterogeneous cell-derived membrane vesicles (exosomes) from a prepurified pellet of cell culture conditioned medium on the grid.


Subject(s)
Cryoelectron Microscopy , DNA , Cryoelectron Microscopy/methods , DNA/chemistry , Extracellular Vesicles/chemistry , Humans , Cholesterol/chemistry , Liposomes/chemistry
7.
J Nanobiotechnology ; 22(1): 233, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38725011

ABSTRACT

BACKGROUND: Dry Eye Disease (DED) is a prevalent multifactorial ocular disease characterized by a vicious cycle of inflammation, oxidative stress, and mitochondrial dysfunction on the ocular surface, all of which lead to DED deterioration and impair the patients' quality of life and social functioning. Currently, anti-inflammatory drugs have shown promising efficacy in treating DED; however, such drugs are associated with side effects. The bioavailability of ocular drugs is less than 5% owing to factors such as rapid tear turnover and the presence of the corneal barrier. This calls for investigations to overcome these challenges associated with ocular drug administration. RESULTS: A novel hierarchical action liposome nanosystem (PHP-DPS@INS) was developed in this study. In terms of delivery, PHP-DPS@INS nanoparticles (NPs) overcame the ocular surface transport barrier by adopting the strategy of "ocular surface electrostatic adhesion-lysosomal site-directed escape". In terms of therapy, PHP-DPS@INS achieved mitochondrial targeting and antioxidant effects through SS-31 peptide, and exerted an anti-inflammatory effect by loading insulin to reduce mitochondrial inflammatory metabolites. Ultimately, the synergistic action of "anti-inflammation-antioxidation-mitochondrial function restoration" breaks the vicious cycle associated with DED. The PHP-DPS@INS demonstrated remarkable cellular uptake, lysosomal escape, and mitochondrial targeting in vitro. Targeted metabolomics analysis revealed that PHP-DPS@INS effectively normalized the elevated level of mitochondrial proinflammatory metabolite fumarate in an in vitro hypertonic model of DED, thereby reducing the levels of key inflammatory factors (IL-1ß, IL-6, and TNF-α). Additionally, PHP-DPS@INS strongly inhibited reactive oxygen species (ROS) production and facilitated mitochondrial structural repair. In vivo, the PHP-DPS@INS treatment significantly enhanced the adhesion duration and corneal permeability of the ocular surface in DED mice, thereby improving insulin bioavailability. It also restored tear secretion, suppressed ocular surface damage, and reduced inflammation in DED mice. Moreover, it demonstrated favorable safety profiles both in vitro and in vivo. CONCLUSION: In summary, this study successfully developed a comprehensive DED management nanosystem that overcame the ocular surface transmission barrier and disrupted the vicious cycle that lead to dry eye pathogenesis. Additionally, it pioneered the regulation of mitochondrial metabolites as an anti-inflammatory treatment for ocular conditions, presenting a safe, efficient, and innovative therapeutic strategy for DED and other inflammatory diseases.


Subject(s)
Dry Eye Syndromes , Inflammation , Liposomes , Mitochondria , Oxidative Stress , Dry Eye Syndromes/drug therapy , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Oxidative Stress/drug effects , Liposomes/chemistry , Inflammation/drug therapy , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Nanoparticles/chemistry , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cornea/metabolism , Cornea/drug effects , Drug Delivery Systems , Oligopeptides
8.
J Nanobiotechnology ; 22(1): 257, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755645

ABSTRACT

Imperceptible examination and unideal treatment effect are still intractable difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). At present, despite 5-fluorouracil (5-FU), as a clinical first-line FOLFIRINOX chemo-drug, has achieved significant therapeutic effects. Nevertheless, these unavoidable factors such as low solubility, lack of biological specificity and easy to induce immunosuppressive surroundings formation, severely limit their treatment in PDAC. As an important source of energy for many tumor cells, tryptophan (Trp), is easily degraded to kynurenine (Kyn) by indolamine 2,3- dioxygenase 1 (IDO1), which activates the axis of Kyn-AHR to form special suppressive immune microenvironment that promotes tumor growth and metastasis. However, our research findings that 5-FU can induce effectively immunogenic cell death (ICD) to further treat tumor by activating immune systems, while the secretion of interferon-γ (IFN-γ) re-induce the Kyn-AHR axis activation, leading to poor treatment efficiency. Therefore, a metal matrix protease-2 (MMP-2) and endogenous GSH dual-responsive liposomal-based nanovesicle, co-loading with 5-FU (anti-cancer drug) and NLG919 (IDO1 inhibitor), was constructed (named as ENP919@5-FU). The multifunctional ENP919@5-FU can effectively reshape the tumor immunosuppression microenvironment to enhance the effect of chemoimmunotherapy, thereby effectively inhibiting cancer growth. Mechanistically, PDAC with high expression of MMP-2 will propel the as-prepared nanovesicle to dwell in tumor region via shedding PEG on the nanovesicle surface, effectively enhancing tumor uptake. Subsequently, the S-S bond containing nanovesicle was cut via high endogenous GSH, leading to the continued release of 5-FU and NLG919, thereby enabling circulating chemoimmunotherapy to effectively cause tumor ablation. Moreover, the combination of ENP919@5-FU and PD-L1 antibody (αPD-L1) showed a synergistic anti-tumor effect on the PDAC model with abdominal cavity metastasis. Collectively, ENP919@5-FU nanovesicle, as a PDAC treatment strategy, showed excellent antitumor efficacy by remodeling tumor microenvironment to circulate tumor chemoimmunotherapy amplification, which has promising potential in a precision medicine approach.


Subject(s)
Carcinoma, Pancreatic Ductal , Fluorouracil , Immunotherapy , Tumor Microenvironment , Tumor Microenvironment/drug effects , Animals , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Mice , Humans , Immunotherapy/methods , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Matrix Metalloproteinase 2/metabolism , Liposomes/chemistry , Kynurenine/metabolism , Interferon-gamma/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use
9.
Int J Nanomedicine ; 19: 4411-4427, 2024.
Article in English | MEDLINE | ID: mdl-38774028

ABSTRACT

Background: Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease characterized by synovial inflammation and joint destruction. Despite progress in RA therapy, it remains difficult to achieve long-term remission in RA patients. Phosphodiesterase 3B (Pde3b) is a member of the phosphohydrolyase family that are involved in many signal transduction pathways. However, its role in RA is yet to be fully addressed. Methods: Studies were conducted in arthritic DBA/1 mice, a suitable mouse strain for collagen-induced rheumatoid arthritis (CIA), to dissect the role of Pde3b in RA pathogenesis. Next, RNAi-based therapy with Pde3b siRNA-loaded liposomes was assessed in a CIA model. To study the mechanism involved, we investigated the effect of Pde3b knockdown on macrophage polarization and related signaling pathway. Results: We demonstrated that mice with CIA exhibited upregulated Pde3b expression in macrophages. Notably, intravenous administration of liposomes loaded with Pde3b siRNA promoted the macrophage anti-inflammatory program and alleviated CIA in mice, as indicated by the reduced inflammatory response, synoviocyte infiltration, and bone and cartilage erosion. Mechanistic study revealed that depletion of Pde3b increased cAMP levels, by which it enhanced PKA-CREB-C/EBPß pathway to transcribe the expression of anti-inflammatory program-related genes. Conclusion: Our results support that Pde3b is involved in the pathogenesis of RA, and Pde3b siRNA-loaded liposomes might serve as a promising therapeutic approach against RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Cyclic Nucleotide Phosphodiesterases, Type 3 , Genetic Therapy , Liposomes , Macrophages , Mice, Inbred DBA , RNA, Small Interfering , Animals , Liposomes/chemistry , Liposomes/administration & dosage , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/therapy , Arthritis, Rheumatoid/chemically induced , Mice , Arthritis, Experimental/genetics , Arthritis, Experimental/prevention & control , Arthritis, Experimental/therapy , Macrophages/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/administration & dosage , Genetic Therapy/methods , Male , Signal Transduction/drug effects
10.
Int J Nanomedicine ; 19: 4377-4409, 2024.
Article in English | MEDLINE | ID: mdl-38774029

ABSTRACT

Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.


Subject(s)
Ischemic Stroke , Nanoparticles , Neovascularization, Physiologic , Humans , Ischemic Stroke/drug therapy , Animals , Nanoparticles/chemistry , Neovascularization, Physiologic/drug effects , Blood-Brain Barrier/drug effects , Liposomes/chemistry , Drug Delivery Systems/methods , Nanoparticle Drug Delivery System/chemistry , Angiogenesis
11.
Biochim Biophys Acta Biomembr ; 1866(5): 184331, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718958

ABSTRACT

The causative genes for neurodegenerative polyglutamine (polyQ) diseases produce homopolymeric polyglutamine (polyQ), polyserine (polyS), polyalanine (polyA), polycysteine (polyC), and polyleucine (polyL) sequences by repeat-associated non-AUG (RAN) translation. The cytotoxicity of the intracellular polyQ and RAN products has been extensively investigated. However, little is known about the toxicity of the extracellular polyQ and RAN products on the membranes of viable cells. Because polyQ aggregates induce a deflated morphology of a model membrane, we hypothesized that extracellular polyQ and RAN products might affect the membrane properties of viable cells. In this study, we demonstrated that exogenous polyS fibrils but not polyS or polyQ non-fibril aggregates altered the thermal phase transition behavior of a model membrane composed of a phosphatidylcholine bilayer using differential scanning calorimetry. PolyS fibrils induced morphological changes in viable red blood cells (RBCs). However, both polyS and polyQ non-fibril aggregates had no effects on RBCs. These results highlight the possibility that extracellular fibrils generated from RAN products may alter the properties of neuronal cell membranes, which may contribute to changes in the brain pathology.


Subject(s)
Erythrocytes , Liposomes , Peptides , Phosphatidylcholines , Erythrocytes/drug effects , Erythrocytes/metabolism , Phosphatidylcholines/chemistry , Humans , Liposomes/chemistry , Peptides/chemistry , Peptides/pharmacology , Cell Membrane/metabolism , Cell Membrane/drug effects , Cell Membrane/chemistry , Phase Transition , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism
12.
ACS Nano ; 18(20): 12737-12748, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38717305

ABSTRACT

Lipids are key factors in regulating membrane fusion. Lipids are not only structural components to form membranes but also active catalysts for vesicle fusion and neurotransmitter release, which are driven by soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. SNARE proteins seem to be partially assembled before fusion, but the mechanisms that arrest vesicle fusion before Ca2+ influx are still not clear. Here, we show that phosphatidylinositol 4,5-bisphosphate (PIP2) electrostatically triggers vesicle fusion as an electrostatic catalyst by lowering the hydration energy and that a myristoylated alanine-rich C-kinase substrate (MARCKS), a PIP2-binding protein, arrests vesicle fusion in a vesicle docking state where the SNARE complex is partially assembled. Vesicle-mimicking liposomes fail to reproduce vesicle fusion arrest by masking PIP2, indicating that native vesicles are essential for the reconstitution of physiological vesicle fusion. PIP2 attracts cations to repel water molecules from membranes, thus lowering the hydration energy barrier.


Subject(s)
Membrane Fusion , Phosphatidylinositol 4,5-Diphosphate , Static Electricity , Water , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Water/chemistry , Liposomes/chemistry , SNARE Proteins/metabolism , SNARE Proteins/chemistry , Catalysis
13.
ACS Nano ; 18(20): 13249-13265, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38720584

ABSTRACT

The therapeutic application of mesenchymal stem cells (MSCs) has good potential as a treatment strategy for systemic lupus erythematosus (SLE), but traditional MSC therapy still has limitations in effectively modulating immune cells. Herein, we present a promising strategy based on dexamethasone liposome-integrated MSCs (Dexlip-MSCs) for treating SLE via multiple immunomodulatory pathways. This therapeutic strategy prolonged the circulation time of dexamethasone liposomes in vivo, restrained CD4+T-cell proliferation, and inhibited the release of proinflammatory mediators (IFN-γ and TNF-α) by CD4+T cells. In addition, Dexlip-MSCs initiated cellular reprogramming by activating the glucocorticoid receptor (GR) signaling pathway to upregulate the expression of anti-inflammatory factors such as cysteine-rich secretory protein LCCL-containing domain 2 (CRISPLD2) and downregulate the expression of proinflammatory factors. In addition, Dexlip-MSCs synergistically increased the anti-inflammatory inhibitory effect of CD4+T cells through the release of dexamethasone liposomes or Dex-integrated MSC-derived exosomes (Dex-MSC-EXOs). Based on these synergistic biological effects, we demonstrated that Dexlip-MSCs alleviated disease progression in MRL/lpr mice more effectively than Dexlip or MSCs alone. These features indicate that our stem cell delivery strategy is a promising therapeutic approach for clinical SLE treatment.


Subject(s)
Dexamethasone , Lupus Erythematosus, Systemic , Mesenchymal Stem Cells , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Dexamethasone/pharmacology , Dexamethasone/chemistry , Lupus Erythematosus, Systemic/therapy , Lupus Erythematosus, Systemic/immunology , Mice , Liposomes/chemistry , Mesenchymal Stem Cell Transplantation , Cell Proliferation/drug effects , Female , Mice, Inbred MRL lpr , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry
14.
Nanoscale ; 16(20): 9836-9852, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38713132

ABSTRACT

Cancer is the second leading cause of death globally after heart diseases. Currently used highly cytotoxic anti-cancer drugs not only kill cancer cells but also often kill non-cancerous healthy body cells, causing adverse side effects. Efforts are now being directed towards developing tumor-selective chemotherapy. Tumor/tumor endothelial cell selective peptide ligands are being covalently grafted onto the exo-surfaces of drug carriers such as liposomes, polymers, etc. A number of prior studies used conjugation of tumor/tumor endothelial cell-selective RGDK- or CGKRK-peptide ligands on the outer surfaces of liposomes, metal-based nanoparticles, single walled carbon nanotubes (SWNTs), etc. However, studies aimed at examining the relative cell membrane fusogenicities and the relative degrees of cellular uptake for the RGDK- and CGKRK-ligand-grafted nanometric drug carriers have not yet been undertaken. Herein, using the widely used liposomes of DOPC, DOPE, DOPS and cholesterol (45 : 25 : 20 : 15, w/w ratio) as the model biomembranes and the fluorescence resonance energy transfer (FRET) assay for measuring membrane fusogenicities, we show that the liposomes of the RGDK-lipopeptide are more biomembrane fusogenic than the liposomes of the CGKRK-lipopeptide. Notably, such FRET assay-derived relative biomembrane fusogenicities of the liposomes of RGDK- and CGKRK-lipopeptides were found to be consistent with their relative degrees of cellular uptake in cultured cancer cells. The present findings open the door for undertaking in-depth in vivo studies aimed at evaluating the relative therapeutic potential of different nanocarriers of drugs/genes/siRNA having tumor-targeting RGDK- and CGKRK-peptides on their exo-surfaces.


Subject(s)
Liposomes , Liposomes/chemistry , Humans , Lipopeptides/chemistry , Lipopeptides/pharmacology , Oligopeptides/chemistry , Cell Membrane/metabolism , Cell Membrane/chemistry , Fluorescence Resonance Energy Transfer , Drug Carriers/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Cholesterol/chemistry , Cholesterol/metabolism , Phosphatidylcholines/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
15.
ACS Appl Mater Interfaces ; 16(20): 25788-25798, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38716694

ABSTRACT

Phototherapy, represented by photodynamic therapy (PDT) and photothermal therapy (PTT), has great potential in tumor treatment. However, the presence of antioxidant glutathione (GSH) and the heat shock proteins (HSPs) expression caused by high temperature can weaken the effects of PDT and PTT. Here, a multifunctional nanocomplex BT&GA@CL is constructed to realize enhanced synergistic PDT/PTT. Cinnamaldehyde liposomes (CLs) formed by cinnamaldehyde dimer self-assembly were loaded with in gambogic acid (GA) and an aggregation-induced emission molecule BT to obtain BT&GA@CL. As a drug carrier, CL can consume glutathione (GSH) and release drugs responsively. The released BT aggregates can simultaneously act as both a photothermal agent and photosensitizer to achieve PDT and PTT under 660 nm laser irradiation. Specifically, GA as an HSP90 inhibitor can attenuate PTT-induced HSP90 protein expression, thereby weakening the tolerance of tumor cells to high temperatures and enhancing PTT. Such a multifunctional nanocomplex simultaneously modulates the content of GSH and HSP90 in tumor cells, thus enhancing both PDT and PTT, ultimately achieving the goal of efficient combined tumor suppression.


Subject(s)
Glutathione , Liposomes , Photochemotherapy , Photosensitizing Agents , Xanthones , Liposomes/chemistry , Glutathione/metabolism , Glutathione/chemistry , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Xanthones/chemistry , Xanthones/pharmacology , Animals , Mice , Photothermal Therapy , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/metabolism , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
16.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732135

ABSTRACT

Glioblastoma (GBM) is the most lethal and common malignant primary brain tumor in adults. An important feature that supports GBM aggressiveness is the unique composition of its extracellular matrix (ECM). Particularly, fibronectin plays an important role in cancer cell adhesion, differentiation, proliferation, and chemoresistance. Thus, herein, a hydrogel with mechanical properties compatible with the brain and the ability to disrupt the dynamic and reciprocal interaction between fibronectin and tumor cells was produced. High-molecular-weight hyaluronic acid (HMW-HA) functionalized with the inhibitory fibronectin peptide Arg-Gly-Asp-Ser (RGDS) was used to produce the polymeric matrix. Liposomes encapsulating doxorubicin (DOX) were also included in the hydrogel to kill GBM cells. The resulting hydrogel containing liposomes with therapeutic DOX concentrations presented rheological properties like a healthy brain. In vitro assays demonstrated that unmodified HMW-HA hydrogels only caused GBM cell killing after DOX incorporation. Conversely, RGDS-functionalized hydrogels displayed per se cytotoxicity. As GBM cells produce several proteolytic enzymes capable of disrupting the peptide-HA bond, we selected MMP-2 to illustrate this phenomenon. Therefore, RGDS internalization can induce GBM cell apoptosis. Importantly, RGDS-functionalized hydrogel incorporating DOX efficiently damaged GBM cells without affecting astrocyte viability, proving its safety. Overall, the results demonstrate the potential of the RGDS-functionalized hydrogel to develop safe and effective GBM treatments.


Subject(s)
Doxorubicin , Fibronectins , Glioblastoma , Hyaluronic Acid , Hydrogels , Oligopeptides , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Fibronectins/metabolism , Fibronectins/antagonists & inhibitors , Hydrogels/chemistry , Cell Line, Tumor , Hyaluronic Acid/chemistry , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Liposomes/chemistry , Apoptosis/drug effects , Matrix Metalloproteinase 2/metabolism
17.
ACS Appl Mater Interfaces ; 16(19): 24295-24307, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38697643

ABSTRACT

Pyroptosis has garnered increasing attention because of its ability to trigger robust antitumor immunity. Pyroptosis is initiated by the activation of inflammasomes, which are regulated by various organelles. The collaboration among organelles offers several protective mechanisms to prevent activation of the inflammasome, thereby limiting the induction of efficient pyroptosis. Herein, a multiorganelle homeostasis disruptor (denoted BLL) is constructed by encapsulating liposomes and bortezomib (BTZ) within a layered double hydroxide (LDH) nanocage to continuously activate inflammasomes for inducing efficient pyroptosis. In lysosomes, the negatively charged liposomes are released to recruit the NLRP3 inflammasomes through electrostatic interactions. ER stress is induced by BTZ to enhance the activation of the NLRP3 inflammasome. Meanwhile, the BLL nanocage exhibited H+-scavenging ability due to the weak alkalinity of LDH, thus disrupting the homeostasis of the lysosome and alleviating the degradation of the NLRP3 inflammasome by lysosomal-associated autophagy. Our results suggest that the BLL nanocage induces homeostatic imbalance in various organelles and efficient pyroptosis. We hope this work can provide new insights into the design of an efficient pyroptosis inducer by disrupting the homeostatic balance of multiple organelles and promote the development of novel antineoplastic platforms.


Subject(s)
Homeostasis , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Pyroptosis/drug effects , Inflammasomes/metabolism , Inflammasomes/drug effects , Homeostasis/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Humans , Mice , Bortezomib/pharmacology , Bortezomib/chemistry , Liposomes/chemistry , Animals , Lysosomes/metabolism , Lysosomes/drug effects , Hydroxides/chemistry , Hydroxides/pharmacology , Nanostructures/chemistry , Nanoparticles/chemistry
18.
J Nanobiotechnology ; 22(1): 299, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38812031

ABSTRACT

BACKGROUND: Discrepancies in the utilization of reactive oxygen species (ROS) between cancer cells and their normal counterparts constitute a pivotal juncture for the precise treatment of cancer, delineating a noteworthy trajectory in the field of targeted therapies. This phenomenon is particularly conspicuous in the domain of nano-drug precision treatment. Despite substantial strides in employing nanoparticles to disrupt ROS for cancer therapy, current strategies continue to grapple with challenges pertaining to efficacy and specificity. One of the primary hurdles lies in the elevated levels of intracellular glutathione (GSH). Presently, predominant methods to mitigate intracellular GSH involve inhibiting its synthesis or promoting GSH efflux. However, a conspicuous gap remains in the absence of a strategy capable of directly and efficiently clearing GSH. METHODS: We initially elucidated the chemical mechanism underpinning oridonin, a diminutive pharmacological agent demonstrated to perturb reactive oxygen species, through its covalent interaction with glutathione. Subsequently, we employed the incorporation of maleimide-liposomes, renowned for their capacity to disrupt the ROS delivery system, to ameliorate the drug's water solubility and pharmacokinetics, thereby enhancing its ROS-disruptive efficacy. In a pursuit to further refine the targeting for acute myeloid leukemia (AML), we harnessed the maleic imide and thiol reaction mechanism, facilitating the coupling of Toll-like receptor 2 (TLR2) peptides to the liposomes' surface via maleic imide. This strategic approach offers a novel method for the precise removal of GSH, and its enhancement endeavors are directed towards fortifying the precision and efficacy of the drug's impact on AML targets. RESULTS: We demonstrated that this peptide-liposome-small molecule machinery targets AML and consequently induces cell apoptosis both in vitro and in vivo through three disparate mechanisms: (I) Oridonin, as a Michael acceptor molecule, inhibits GSH function through covalent bonding, triggering an initial imbalance of oxidative stress. (II) Maleimide further induces GSH exhaustion, aggravating redox imbalance as a complementary augment with oridonin. (III) Peptide targets TLR2, enhances the directivity and enrichment of oridonin within AML cells. CONCLUSION: The rationally designed nanocomplex provides a ROS drug enhancement and targeted delivery platform, representing a potential solution by disrupting redox balance for AML therapy.


Subject(s)
Diterpenes, Kaurane , Glutathione , Leukemia, Myeloid, Acute , Liposomes , Reactive Oxygen Species , Diterpenes, Kaurane/chemistry , Diterpenes, Kaurane/pharmacology , Glutathione/metabolism , Glutathione/chemistry , Liposomes/chemistry , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Humans , Reactive Oxygen Species/metabolism , Animals , Mice , Cell Line, Tumor , Toll-Like Receptor 2/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects
19.
Mol Pharm ; 21(6): 2970-2980, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38742943

ABSTRACT

One of the most significant reasons hindering the clinical translation of nanomedicines is the rapid clearance of intravenously injected nanoparticles by the mononuclear phagocyte system, particularly by Kupffer cells in the liver, leading to an inefficient delivery of nanomedicines for tumor treatment. The threshold theory suggests that the liver's capacity to clear nanoparticles is limited, and a single high dose of nanoparticles can reduce the hepatic clearance efficiency, allowing more nanomedicines to reach tumor tissues and enhance therapeutic efficacy. Building upon this theory, researchers have conducted numerous validation studies based on the same nanoparticle carrier systems. These studies involve the use of albumin nanoparticles to improve the therapeutic efficacy of albumin nanomedicines as well as polyethylene glycol (PEG)-modified liposomal nanoparticles to enhance the efficacy of PEGylated liposomal nanomedicines. However, there is no research indicating the feasibility of the threshold theory when blank nanoparticles and nanomedicine belong to different nanoparticle carrier systems currently. In this study, we prepared two different sizes of albumin nanoparticles by using bovine serum albumin. We used the marketed nanomedicine liposomal doxorubicin hydrochloride injection (trade name: LIBOD, manufacturer: Shanghai Fudan-zhangjiang Biopharmaceutical Co., Ltd.), as the representative nanomedicine. Through in vivo experiments, we found that using threshold doses of albumin nanoparticles still can reduce the clearance rate of LIBOD, prolong its time in vivo, increase the area under the plasma concentration-time curve (AUC), and also lead to an increased accumulation of the drug at the tumor site. Furthermore, evaluation of in vivo efficacy and safety further indicates that threshold doses of 100 nm albumin nanoparticles can enhance the antitumor effect of LIBOD without causing harm to the animals. During the study, we found that the particle size of albumin nanoparticles influenced the in vivo distribution of the nanomedicine at the same threshold dose. Compared with 200 nm albumin nanoparticles, 100 nm albumin nanoparticles more effectively reduce the clearance efficiency of LIBOD and enhance nanomedicine accumulation at the tumor site, warranting further investigation. This study utilized albumin nanoparticles to reduce hepatic clearance efficiency and enhance the delivery efficiency of nonalbumin nanocarrier liposomal nanomedicine, providing a new avenue to improve the efficacy and clinical translation of nanomedicines with different carrier systems.


Subject(s)
Doxorubicin , Nanoparticles , Polyethylene Glycols , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Animals , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Mice , Liposomes/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/administration & dosage , Tissue Distribution , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Mice, Inbred BALB C , Liver/drug effects , Liver/metabolism , Particle Size , Nanomedicine/methods , Humans , Male , Female
20.
Biomater Adv ; 161: 213891, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38781738

ABSTRACT

An antitumour chemo-photodynamic therapy nanoplatform was constructed based on phospholipid-coated NaYF4: Yb/Er upconversion nanoparticles (UCNPs). In this work, the amphiphilic block copolymer DSPE-PEG2000 was combined with the surface ligand oleic acid of the UCNPs through hydrophobic interaction to form liposomes with a dense hydrophobic layer in which the photosensitizer hypocrellin B (HB) was assembled. The coated HB formed J-aggregates, which caused a large redshift in the absorption spectrum and improved the quantum efficiency of energy transfer. Furthermore, MnO2 nanosheets grew in-situ on the liposomes through OMn coordination. Therefore, a multifunctional tumour microenvironment (TME)-responsive theranostic nanoplatform integrating photodynamic therapy (PDT) and chemodynamic therapy (CDT) was successfully developed. The results showed that this NIR-mediated chemo-photodynamic therapy nanoplatform was highly efficient for oncotherapy.


Subject(s)
Manganese Compounds , Nanoparticles , Oxides , Perylene , Photochemotherapy , Photosensitizing Agents , Quinones , Photochemotherapy/methods , Perylene/analogs & derivatives , Perylene/pharmacology , Perylene/chemistry , Perylene/administration & dosage , Humans , Quinones/chemistry , Quinones/pharmacology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Oxides/chemistry , Oxides/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/administration & dosage , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Animals , Phenol/chemistry , Phenol/pharmacology , Liposomes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Mice , Cell Line, Tumor , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...