Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51.889
Filter
1.
Food Chem ; 462: 141008, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39217746

ABSTRACT

Hydrophobic bioactive compounds like astaxanthin (AST) exhibit poor water solubility and low bioavailability. Liposomes, which serve as nanocarriers, are known for their excellent biocompatibility and minimal immunogenicity. Traditionally, liposomes have been primarily constructed using phospholipids and cholesterol. However, the intake of cholesterol may pose a risk to human health. Phytosterol ester was reported to reduce level of cholesterol and improve properties of liposomes. In this study, phytosterol oleate was used to prepare liposomes instead of cholesterol to deliver AST (AST-P-Lip). The size range of AST-P-Lip was 100-220 nm, and the morphology was complete and uniform. In vitro studies showed that AST-P-Lip significantly enhanced the antioxidant activity and oral bioavailability of AST. During simulated digestion, AST-P-Lip protected AST from damage by gastric and intestinal digestive fluid. Additionally, AST-P-Lip had a good storage stability and safety. These results provide references for the preparation of novel liposomes and the delivery of bioactive compounds.


Subject(s)
Cholesterol , Liposomes , Phytosterols , Xanthophylls , Liposomes/chemistry , Xanthophylls/chemistry , Xanthophylls/pharmacology , Xanthophylls/administration & dosage , Humans , Phytosterols/chemistry , Phytosterols/pharmacology , Phytosterols/administration & dosage , Cholesterol/chemistry , Particle Size , Biological Availability , Oleic Acid/chemistry , Drug Compounding , Animals , Antioxidants/chemistry , Antioxidants/pharmacology
2.
Biomaterials ; 313: 122801, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39236630

ABSTRACT

Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.


Subject(s)
Immunotherapy , Liposomes , Neoplasms , Liposomes/chemistry , Humans , Immunotherapy/methods , Animals , Neoplasms/therapy , Neoplasms/drug therapy , Drug Delivery Systems/methods , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage
3.
Biomaterials ; 312: 122712, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39098305

ABSTRACT

Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.


Subject(s)
Liposomes , MicroRNAs , Photochemotherapy , Photosensitizing Agents , Triple Negative Breast Neoplasms , Tumor Microenvironment , Animals , Humans , Liposomes/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Photochemotherapy/methods , Tumor Microenvironment/drug effects , Cell Line, Tumor , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Female , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Mice , Aptamers, Nucleotide/chemistry , Delayed-Action Preparations/chemistry , RNA Interference , Zebrafish
4.
Biomaterials ; 312: 122747, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39142219

ABSTRACT

Directly administering medication to inflamed intestinal sites for treating ulcerative colitis (UC), poses significant challenges like retention time, absorption variability, side effects, drug stability, and non-specific delivery. Recent advancements in therapy to treat colitis aim to improve local drug availability that is enema therapy at the site of inflammation, thereby reducing systemic adverse effects. Nevertheless, a key limitation lies in enemas' inability to sustain medication in the colon due to rapid peristaltic movement, diarrhea, and poor local adherence. Therefore, in this work, we have developed site-specific thiolated mucoadhesive anionic nanoliposomes to overcome the limitations of conventional enema therapy. The thiolated delivery system allows prolonged residence of the delivery system at the inflamed site in the colon, confirmed by the adhesion potential of thiolated nanoliposomes using in-vitro and in-vivo models. To further provide therapeutic efficacy thiolated nanoliposomes were loaded with gallic acid (GA), a natural compound known for its antibacterial, antioxidant, and potent anti-inflammatory properties. Consequently, Gallic Acid-loaded Thiolated 2,6 DALP DMPG (GATh@APDL) demonstrates the potential for targeted adhesion to the inflamed colon, facilitated by their small size 100 nm and anionic nature. Therapeutic studies indicate that this formulation offers protective effects by mitigating colonic inflammation, downregulating the expression of NF-κB, HIF-1α, and MMP-9, and demonstrating superior efficacy compared to the free GA enema. The encapsulated GA inhibits the NF-κB expression, leading to enhanced expression of MUC2 protein, thereby promoting mucosal healing in the colon. Furthermore, GATh@APDL effectively reduces neutrophil infiltration and regulates immune cell quantification in colonic lamina propria. Our findings suggest that GATh@APDL holds promise for alleviating UC and addressing the limitations of conventional enema therapy.


Subject(s)
Colitis, Ulcerative , Liposomes , Sulfhydryl Compounds , Colitis, Ulcerative/drug therapy , Liposomes/chemistry , Animals , Sulfhydryl Compounds/chemistry , Humans , Nanoparticles/chemistry , Mice , Colon/pathology , Colon/drug effects , Colon/metabolism , Male , Drug Delivery Systems
5.
AAPS PharmSciTech ; 25(7): 208, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237678

ABSTRACT

Mathematical modeling of drug release from drug delivery systems is crucial for understanding and optimizing formulations. This research provides a comparative mathematical analysis of drug release from lipid-based nanoparticles. Drug release profiles from various types of lipid nanoparticles, including liposomes, nanostructured lipid carriers (NLCs), solid lipid nanoparticles (SLNs), and nano/micro-emulsions (NEMs/MEMs), were extracted from the literature and used to assess the suitability of eight conventional mathematical release models. For each dataset, several metrics were calculated, including the coefficient of determination (R2), adjusted R2, the number of errors below certain thresholds (5%, 10%, 12%, and 20%), Akaike information criterion (AIC), regression sum square (RSS), regression mean square (RMS), residual sum of square (rSS), and residual mean square (rMS). The Korsmeyer-Peppas model ranked highest among the evaluated models, with the highest adjusted R2 values of 0.95 for NLCs and 0.93 for other liposomal drug delivery systems. The Weibull model ranked second, with adjusted R2 values of 0.92 for liposomal systems, 0.94 for SLNs, and 0.82 for NEMs/MEMs. Thus, these two models appear to be more effective in forecasting and characterizing the release of lipid nanoparticle drugs, potentially making them more suitable for upcoming research endeavors.


Subject(s)
Drug Delivery Systems , Drug Liberation , Lipids , Liposomes , Nanoparticles , Nanoparticles/chemistry , Lipids/chemistry , Liposomes/chemistry , Drug Delivery Systems/methods , Models, Theoretical , Drug Carriers/chemistry , Emulsions/chemistry , Chemistry, Pharmaceutical/methods
6.
Vet Med Sci ; 10(5): e70017, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39239721

ABSTRACT

BACKGROUND: Nuciferine (NUC), a natural compound extracted from lotus leaves, has been proven to have anti-obesity effects. However, the development and application of NUC as an anti-obesity drug in dogs are hindered due to its poor water solubility and low bioavailability. OBJECTIVE: To promote the development of NUC-related products for anti-obesity in dogs, this study prepared NUC into a liposome formulation and evaluated its characteristics, pharmacokinetics in dogs, and anti-obesity effects on high-fat diet dogs. METHODS: NUC liposomes were prepared by the ethanol injection method, using NUC, egg lecithin, and ß-sitosterol as raw materials. The characteristics and release rate in vitro of liposomes were evaluated by particle size analyser and dialysis method, respectively. The pharmacokinetics in dogs after oral administration of NUC-liposomes was carried out by the high-performance liquid chromatography (HPLC) method. Moreover, we investigated the anti-obesity effect of NUC-liposomes on obese dogs fed with a high-fat diet. RESULTS: NUC-liposome was successfully prepared, with an EE of (79.31 ± 1.06)%, a particle size of (81.25 ± 3.14) nm, a zeta potential of (-18.75 ± 0.23) mV, and a PDI of 0.175 ± 0.031. The cumulative release rate in vitro of NUC from NUC-liposomes was slower than that of NUC. The T1/2 and relative bioavailability of NUC-liposomes in dogs increased, and CL reduced compared with NUC. In addition, the preventive effect of NUC-liposomes on obesity in high-fat diet dogs is stronger than that of NUC. CONCLUSIONS: The liposome formulation of NUC was conducive to improve its relative bioavailability and anti-obesity effect in dogs.


Subject(s)
Anti-Obesity Agents , Aporphines , Liposomes , Obesity , Animals , Dogs , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/chemistry , Obesity/veterinary , Obesity/drug therapy , Male , Aporphines/pharmacokinetics , Aporphines/chemistry , Aporphines/administration & dosage , Diet, High-Fat , Dog Diseases/drug therapy , Dog Diseases/prevention & control , Female
7.
Int J Nanomedicine ; 19: 8949-8970, 2024.
Article in English | MEDLINE | ID: mdl-39246424

ABSTRACT

Introduction: RNA interference (RNAi) stands as a widely employed gene interference technology, with small interfering RNA (siRNA) emerging as a promising tool for cancer treatment. However, the inherent limitations of siRNA, such as easy degradation and low bioavailability, hamper its efficacy in cancer therapy. To address these challenges, this study focused on the development of a nanocarrier system (HLM-N@DOX/R) capable of delivering both siRNA and doxorubicin for the treatment of breast cancer. Methods: The study involved a comprehensive investigation into various characteristics of the nanocarrier, including shape, diameter, Fourier transform infrared (FT-IR) spectroscopy, X-ray photoelectron spectroscopy (XPS), encapsulation efficiency, and drug loading. Subsequently, in vitro and in vivo studies were conducted on cytotoxicity, cellular uptake, cellular immunofluorescence, lysosome escape, and mouse tumor models to evaluate the efficacy of the nanocarrier in reversing tumor multidrug resistance and anti-tumor effects. Results: The results showed that HLM-N@DOX/R had a high encapsulation efficiency and drug loading capacity, and exhibited pH/redox dual responsive drug release characteristics. In vitro and in vivo studies showed that HLM-N@DOX/R inhibited the expression of P-gp by 80%, inhibited MDR tumor growth by 71% and eliminated P protein mediated multidrug resistance. Conclusion: In summary, HLM-N holds tremendous potential as an effective and targeted co-delivery system for DOX and P-gp siRNA, offering a promising strategy for overcoming MDR in breast cancer.


Subject(s)
Breast Neoplasms , Doxorubicin , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Liposomes , RNA, Small Interfering , Animals , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/administration & dosage , Female , Liposomes/chemistry , Mice , Drug Resistance, Neoplasm/drug effects , Humans , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacokinetics , Drug Resistance, Multiple/drug effects , Breast Neoplasms/drug therapy , Cell Line, Tumor , MCF-7 Cells , Mice, Inbred BALB C , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Nanoparticles/chemistry , Drug Liberation , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Xenograft Model Antitumor Assays
8.
Int J Nanomedicine ; 19: 8929-8947, 2024.
Article in English | MEDLINE | ID: mdl-39246429

ABSTRACT

Introduction: Cell death regulation holds a unique value in the field of cancer therapy. Recently, disulfidptosis has garnered substantial scientific attention. Previous studies have reported that sonodynamic therapy (SDT) based on reactive oxygen species (ROS) can regulate cancer cell death, achieving an limited anti-cancer effect. However, the integration of SDT with disulfidptosis as an anti-cancer strategy has not been extensively developed. In this study, we constructed an artificial membrane disulfidptosis sonosensitizer, specifically, a nanoliposome (SC@lip) coated with a combination of the chemotherapy medicine Sorafenib (Sora) and sonosensitizer Chlorin e6 (Ce6), to realize a one-stop enhanced SDT effect that induces disulfidptosis-like cancer cell death. Methods: Sorafenib and Ce6 were co-encapsulated into PEG-modified liposomes, and SC@Lip was constructed using a simple rotary evaporation phacoemulsification method. The cell phagocytosis, ROS generation ability, glutathione (GSH) depletion ability, lipid peroxidation (LPO), and disulfidptosis-like death mediated by SC@Lip under ultrasound (US) irradiation were evaluated. Based on a 4T1 subcutaneous tumor model, both the in vivo biological safety assessment and the efficacy of SDT were assessed. Results: SC@Lip exhibits high efficiency in cellular phagocytosis. After being endocytosed by 4T1 cells, abundant ROS were produced under SDT activation, and the cell survival rates were below 5%. When applied to a 4T1 subcutaneous tumor model, the enhanced SDT mediated by SC@Lip inhibited tumor growth and prolonged the survival time of mice. In vitro and in vivo experiments show that SC@Lip can enhance the SDT effect and trigger disulfidptosis-like cancer cell death, thus achieving anti-tumor efficacy both in vitro and in vivo. Conclusion: SC@Lip is a multifunctional nanoplatform with an artificial membrane, which can integrate the functions of sonosensitization and GSH depletion into a biocompatible nanoplatform, and can be used to enhance the SDT effect and promote disulfidptosis-like cancer cell death.


Subject(s)
Chlorophyllides , Lipid Peroxidation , Liposomes , Porphyrins , Reactive Oxygen Species , Sorafenib , Ultrasonic Therapy , Animals , Liposomes/chemistry , Lipid Peroxidation/drug effects , Sorafenib/pharmacology , Sorafenib/chemistry , Ultrasonic Therapy/methods , Mice , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/administration & dosage , Female , Mice, Inbred BALB C , Nanoparticles/chemistry , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Glutathione/metabolism , Cell Death/drug effects
9.
Int J Nanomedicine ; 19: 8769-8778, 2024.
Article in English | MEDLINE | ID: mdl-39220196

ABSTRACT

Introduction: The tumor microenvironment (TME) of pancreatic cancer is highly immunosuppressive and characterized by a large number of cancer-associated fibroblasts, myeloid-derived suppressor cells, and regulatory T cells. Stimulator of interferon genes (STING) is an endoplasmic reticulum receptor that plays a critical role in immunity. STING agonists have demonstrated the ability to inflame the TME, reduce tumor burden, and confer anti-tumor activity in mouse models. 2'3' cyclic guanosine monophosphate adenosine monophosphate (2'3'-cGAMP) is a high-affinity endogenous ligand of STING. However, delivering cGAMP to antigen-presenting cells and tumor cells within the cytosol remains challenging due to membrane impermeability and poor stability. Methods: In this study, we encapsulated 2'3'-cGAMP in a lipid nanoparticle (cGAMP-LNP) designed for efficient cellular delivery. We assessed the properties of the nanoparticles using a series of in-vitro studies designed to evaluate their cellular uptake, cytosolic release, and minimal cytotoxicity. Furthermore, we examined the nanoparticle's anti-tumor effect in a syngeneic mouse model of pancreatic cancer. Results: The lipid platform significantly increased the cellular uptake of 2'3'-cGAMP. cGAMP-LNP exhibited promising antitumor activity in the syngeneic mouse model of pancreatic cancer. Discussion: The LNP platform shows promise for delivering exogenous 2'3'-cGAMP or its derivatives in cancer therapy.


Subject(s)
Membrane Proteins , Nanoparticles , Nucleotides, Cyclic , Pancreatic Neoplasms , Tumor Microenvironment , Animals , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Nucleotides, Cyclic/pharmacology , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/pharmacokinetics , Nucleotides, Cyclic/administration & dosage , Membrane Proteins/agonists , Mice , Cell Line, Tumor , Humans , Tumor Microenvironment/drug effects , Mice, Inbred C57BL , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Liposomes/chemistry , Liposomes/pharmacology , Liposomes/pharmacokinetics , Liposomes/administration & dosage
10.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39240335

ABSTRACT

Predicting the immunogenicity of candidate vaccines in humans remains a challenge. To address this issue, we developed a lymphoid organ-chip (LO chip) model based on a microfluidic chip seeded with human PBMC at high density within a 3D collagen matrix. Perfusion of the SARS-CoV-2 spike protein mimicked a vaccine boost by inducing a massive amplification of spike-specific memory B cells, plasmablast differentiation, and spike-specific antibody secretion. Features of lymphoid tissue, including the formation of activated CD4+ T cell/B cell clusters and the emigration of matured plasmablasts, were recapitulated in the LO chip. Importantly, myeloid cells were competent at capturing and expressing mRNA vectored by lipid nanoparticles, enabling the assessment of responses to mRNA vaccines. Comparison of on-chip responses to Wuhan monovalent and Wuhan/Omicron bivalent mRNA vaccine boosts showed equivalent induction of Omicron neutralizing antibodies, pointing at immune imprinting as reported in vivo. The LO chip thus represents a versatile platform suited to the preclinical evaluation of vaccine-boosting strategies.


Subject(s)
COVID-19 Vaccines , COVID-19 , Memory B Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , mRNA Vaccines , Humans , COVID-19 Vaccines/immunology , mRNA Vaccines/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Memory B Cells/immunology , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Lymphoid Tissue/immunology , Lab-On-A-Chip Devices , Vaccines, Synthetic/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Liposomes , Nanoparticles
11.
J Nanobiotechnology ; 22(1): 551, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252079

ABSTRACT

Chemodynamic therapy represents a novel tumor therapeutic modality via triggering catalytic reactions in tumors to yield highly toxic reactive oxygen species (ROS). Nevertheless, low efficiency catalytic ability, potential systemic toxicity and inefficient tumor targeting, have hindered the efficacy of chemodynamic therapy. Herein, a rationally designed catalytic nanoplatform, composed of folate acid conjugated liposomes loaded with copper peroxide (CP) and chloroquine (CQ; a clinical drug) (denoted as CC@LPF), could power maximal tumor cytotoxicity, mechanistically via maneuvering endogenous and exogenous copper for a highly efficient catalytic reaction. Despite a massive autophagosome accumulation elicited by CP-powered autophagic initiation and CQ-induced autolysosomal blockage, the robust ROS, but not aberrant autophagy, underlies the synergistic tumor inhibition. Otherwise, this combined mode also elicits an early onset, above all, long-term high-level existence of immunogenic cell death markers, associated with ROS and aberrant autophagy -triggered endoplasmic reticulum stress. Besides, CC@LPF, with tumor targeting capability and selective tumor cytotoxicity, could elicit intratumor dendritic cells (mainly attributed to CQ) and tumor infiltrating CD8+ T cells, upon combining with PD-L1 therapeutic antibody, further induce significant anti-tumor effect. Collectively, the rationally designed nanoplatform, CC@LPF, could enhance tumor chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment.


Subject(s)
Copper , Immunotherapy , Reactive Oxygen Species , Tumor Microenvironment , Tumor Microenvironment/drug effects , Animals , Copper/chemistry , Copper/pharmacology , Mice , Immunotherapy/methods , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Liposomes/chemistry , Catalysis , Autophagy/drug effects , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , Chloroquine/pharmacology , Female , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
BMC Res Notes ; 17(1): 239, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223611

ABSTRACT

OBJECTIVES: The Ishikawa cell line is the most widely used model system for investigating implantation and endometrial cancer. Understanding the biology of this cell line is essential for developing effective interventional strategies. To gain a deeper understanding of its cellular protein profile, we extracted cellular proteins from Ishikawa cells and analyzed the peptides using mass spectrometry. Our goal was to create a proteomic resource specifically tailored for Ishikawa cells. This data set is of particular significance in the realm of targeted drug delivery. Liposomes are synthetic spherical vesicles composed of hydrophobic bilayer phospholipids and have received immense recognition as highly effective carriers for the delivery of pharmaceutical drugs and essential nutrients to the endometrium. Phosphatidylcholine and phosphatidylethanolamine are often combined to create functional liposomal systems. To discern any potential interfering effects originating from the liposome backbone, our investigation involved direct effects of phospholipid liposomes on endometrial epithelial cells. DATA DESCRIPTION: The data set includes peptide spectra derived from the intracellular proteomes of Ishikawa endometrial cancer cell isolates and their phospholipid-treated counterparts. Representing a proteome-wide profile, this dataset aims to contribute to a broader understanding of the physiology of endometrial epithelial cells. Proteomic analysis identified key proteins involved in the intricate regulation of cellular metabolism, cell cycle progression, and signaling. Between-group analysis revealed no differentially expressed proteins after adjusting for multiple testing using the applied thresholds (p-value < 0.05 and |logFC| > 1). Data are available via ProteomeXchange with identifier PXD050871.


Subject(s)
Endometrial Neoplasms , Liposomes , Proteomics , Female , Humans , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Proteomics/methods , Cell Line, Tumor , Proteome/metabolism , Phosphatidylethanolamines/metabolism , Phosphatidylcholines/metabolism
13.
AAPS J ; 26(5): 99, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231845

ABSTRACT

ß-site amyloid precursor protein cleaving enzyme (BACE1) represents a key target for Alzheimer's disease (AD) therapy because it is essential for producing the toxic amyloid ß (Aß) peptide that plays a crucial role in the disease's development. BACE1 inhibitors are a promising approach to reducing Aß levels in the brain and preventing AD progression. However, systemic delivery of such inhibitors to the brain demonstrates limited efficacy because of the presence of the blood-brain barrier (BBB). Nose-to-brain (NtB) delivery has the potential to overcome this obstacle. Liposomal drug delivery systems offer several advantages over traditional methods for delivering drugs and nucleic acids from the nose to the brain. The current study aims to prepare, characterize, and evaluate in vitro liposomal forms of donepezil, memantine, BACE-1 siRNA, and their combination for possible treatment of AD via NtB delivery. All the liposomal formulations were prepared using the rotary evaporation method. Their cellular internalization, cytotoxicity, and the suppression of beta-amyloid plaque and other pro-inflammatory cytokine expressions were studied. The Calu-3 Transwell model was used as an in vitro system for mimicking the anatomical and physiological conditions of the nasal epithelium and studying the suitability of the proposed formulations for possible NtB delivery. The investigation results show that liposomes provided the effective intracellular delivery of therapeutics, the potential to overcome tight junctions in BBB, reduced beta-amyloid plaque accumulation and pro-inflammatory cytokine expression, supporting the therapeutic potential of our approach.


Subject(s)
Administration, Intranasal , Alzheimer Disease , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Donepezil , Liposomes , RNA, Small Interfering , Alzheimer Disease/drug therapy , Humans , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Donepezil/administration & dosage , Drug Delivery Systems/methods , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Brain/metabolism , Brain/drug effects , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Piperidines/pharmacology , Nasal Mucosa/metabolism , Nasal Mucosa/drug effects , Indans/administration & dosage , Indans/pharmacokinetics , Amyloid beta-Peptides/metabolism
14.
Adv Food Nutr Res ; 112: 257-300, 2024.
Article in English | MEDLINE | ID: mdl-39218504

ABSTRACT

Natural bioactive compounds with antioxidant, antimicrobial, anticancer, and other biological activities are vital for maintaining the body's physiological functions and enhancing immunity. These compounds have great potential as nutritional therapeutic agents, but they can be limited due to their poor flavor, color, unstable nature, and poor water solubility, and degradation by gastrointestinal enzymes. Liposomes, as ideal carriers, can encapsulate both water-soluble and fat-soluble nutrients, enhance the bioavailability of functional substances, promote the biological activity of functional substances, and control the release of nutrients. Despite their potential, liposomes still face obstacles in nutrient delivery. Therefore, the design of liposomes for special needs, optimization of the liposome preparation process, enhancement of liposome encapsulation efficiency, and industrial production are key issues that must be addressed in order to develop food-grade liposomes. Moreover, the research on surface-targeted modification and surface functionalization of liposomes is valuable for expanding the scope of application of liposomes and achieving the release of functional substances from liposomes at the appropriate time and site. The establishment of in vivo and in vitro digestion models of nutrient-loaded liposomes, in-depth study of gastrointestinal digestive behavior after liposome ingestion, targeted nutrient release, and deciphering the nutritional intervention of human diseases and positive health promotion are promising fields with broad development prospects.


Subject(s)
Liposomes , Humans , Drug Delivery Systems , Biological Availability , Functional Food
15.
J Nanobiotechnology ; 22(1): 553, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261807

ABSTRACT

Lipid nanoparticles (LNPs) are currently the most commonly used non-viral gene delivery system. Their physiochemical attributes, encompassing size, charge and surface modifications, significantly affect their behaviors both in vivo and in vitro. Nevertheless, the effects of these properties on the transfection and distribution of LNPs after intramuscular injection remain elusive. In this study, LNPs with varying sizes, lipid-based charges and PEGylated lipids were formulated to study their transfection and in vivo distribution. Luciferase mRNA (mLuc) was entraped in LNPs as a model nucleic acid molecule. Results indicated that smaller-sized LNPs and those with neutral potential presented superior transfection efficiency after intramuscular injection. Surprisingly, the sizes and charges did not exert a notable influence on the in vivo distribution of the LNPs. Furthermore, PEGylated lipids with shorter acyl chains contributed to enhanced transfection efficiency due to their superior cellular uptake and lysosomal escape capabilities. Notably, the mechanisms underlying cellular uptake differed among LNPs containing various types of PEGylated lipids, which was primarily attributed to the length of their acyl chain. Together, these insights underscore the pivotal role of nanoparticle characteristics and PEGylated lipids in the intramuscular route. This study not only fills crucial knowledge gaps but also provides significant directions for the effective delivery of mRNA via LNPs.


Subject(s)
Lipids , Nanoparticles , Particle Size , Polyethylene Glycols , RNA, Messenger , Transfection , Nanoparticles/chemistry , Animals , Polyethylene Glycols/chemistry , Injections, Intramuscular , Lipids/chemistry , Transfection/methods , Mice , Gene Transfer Techniques , Humans , Luciferases/metabolism , Luciferases/genetics , Surface Properties , Liposomes
16.
AAPS PharmSciTech ; 25(7): 213, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266895

ABSTRACT

To overcome the challenges associated with the co-delivery of AuNPs (gold nanoparticles) and miRNA as an anti-breast cancer combination therapy, niosomal systems were developed using Span 60, cholesterol, and a cationic lipid (CTAB), and the formulations were optimized using Box-Behnken experimental design. The niosomal formulations with the smallest size were selected for further optimization of size, surface charge, entrapment efficiency, and stability. To achieve this, AuNPs and DSPE-PEG2000 (2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000)were added to the formulation. The optimized niosomal formulation could effectively encapsulate AuNPs with an entrapment efficiency of 34.49% ± 0.84 and a spherical particle size of 153.6 ± 4.62 nm. The incorporation of PEG and CTAB led to notable enhancements in the overall characteristics of the delivery system. To evaluate the effectiveness of the combination therapy, various assessments such as cytotoxicity, apoptosis, and gene expression properties were conducted. The results demonstrated that the combination delivery using the new C-PEG-Nio-AuNPs (cationic pegylated niosomal gold nanoparticles) system and miRNA had the lowest IC50, the highest apoptosis rate, and the most significant upregulation of miRNA and BAX/BCL2 expression in MCF-7 cell growth. In conclusion, this innovative co-delivery approach represents a promising breakthrough in the development of therapeutic agents for breast cancer treatment. By combining multiple therapeutic agents within a single delivery system, this method has the potential to enhance treatment efficacy, reduce side effects, and improve patient outcomes.


Subject(s)
Breast Neoplasms , Gold , Liposomes , Metal Nanoparticles , MicroRNAs , Particle Size , Polyethylene Glycols , Gold/chemistry , Humans , MicroRNAs/administration & dosage , MCF-7 Cells , Polyethylene Glycols/chemistry , Metal Nanoparticles/chemistry , Liposomes/chemistry , Breast Neoplasms/drug therapy , Female , Cations/chemistry , Apoptosis/drug effects , Drug Delivery Systems/methods , Cell Survival/drug effects , Phosphatidylethanolamines/chemistry
17.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273099

ABSTRACT

Cholesterol homeostasis is pivotal for cellular function. Acyl-coenzyme A:cholesterol acyltransferase 1 (ACAT1), also abbreviated as SOAT1, is an enzyme responsible for catalyzing the storage of excess cholesterol to cholesteryl esters. ACAT1 is an emerging target to treat diverse diseases including atherosclerosis, cancer, and neurodegenerative diseases. F12511 is a high-affinity ACAT1 inhibitor. Previously, we developed a stealth liposome-based nanoparticle to encapsulate F12511 to enhance its delivery to the brain and showed its efficacy in treating a mouse model for Alzheimer's disease (AD). In this study, we introduce F26, a close derivative of F12511 metabolite in rats. F26 was encapsulated in the same DSPE-PEG2000/phosphatidylcholine (PC) liposome-based nanoparticle system. We employed various in vitro and in vivo methodologies to assess F26's efficacy and toxicity compared to F12511. The results demonstrate that F26 is more effective and durable than F12511 in inhibiting ACAT1, in both mouse embryonic fibroblasts (MEFs), and in multiple mouse tissues including the brain tissues, without exhibiting any overt systemic or neurotoxic effects. This study demonstrates the superior pharmacokinetic and safety profile of F26 in wild-type mice, and suggests its therapeutic potential against various neurodegenerative diseases including AD.


Subject(s)
Liposomes , Nanoparticles , Sterol O-Acyltransferase , Animals , Liposomes/chemistry , Mice , Nanoparticles/chemistry , Sterol O-Acyltransferase/antagonists & inhibitors , Sterol O-Acyltransferase/metabolism , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/metabolism , Brain/metabolism , Brain/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Rats , Male , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism
18.
Int J Mol Sci ; 25(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39273463

ABSTRACT

Phospholipase A2 (PLA2) is a superfamily of phospholipase enzymes that dock at the water/oil interface of phospholipid assemblies, hydrolyzing the ester bond at the sn-2 position. The enzymatic activity of these enzymes differs based on the nature of the substrate, its supramolecular assemblies (micelle, liposomes), and their composition, reflecting the interfacial nature of the PLA2s and requiring assays able to directly quantify this interaction of the enzyme(s) with these supramolecular assemblies. We developed and optimized a simple, universal assay method employing the pH-sensitive indicator dye bromothymol blue (BTB), in which different POPC (3-palmitoyl-2-oleoyl-sn-glycero-1-phosphocholine) self-assemblies (liposomes or mixed micelles with Triton X-100 at different molar ratios) were used to assess the enzymatic activity. We used this assay to perform a comparative analysis of PLA2 kinetics on these supramolecular assemblies and to determine the kinetic parameters of PLA2 isozymes IB and IIA for each supramolecular POPC assembly. This assay is suitable for assessing the inhibition of PLA2s with great accuracy using UV-VIS spectrophotometry, being thus amenable for screening of PLA2 enzymes and their substrates and inhibitors in conditions very similar to physiologic ones.


Subject(s)
Phosphatidylcholines , Phospholipases A2 , Phospholipases A2/metabolism , Phospholipases A2/chemistry , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Kinetics , Micelles , Liposomes/chemistry , Hydrogen-Ion Concentration , Enzyme Assays/methods , Octoxynol/chemistry
19.
Pestic Biochem Physiol ; 204: 106082, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277395

ABSTRACT

Bemisia tabaci poses a severe threat to plants, and the control of B. tabaci mainly relies on pesticides, which causes more and more rapidly increasing resistance. ß-Caryophyllene is a promising ingredient for agricultural pest control, but its feature of poor water solubility need to be improved in practical applications. Nanotechnology can enhance the effectiveness and dispersion of volatile organic compounds (VOCs). In this study, a nanoliposome carrier was constructed by ethanol injection and ultrasonic dispersion method, and ß-caryophyllene was wrapped inside it, thus solving the defect of poor solubility of ß-caryophyllene. The size of the ß-caryophyllene nanoliposomes (C-BT-NPs) was around 200 nm, with the absolute value of the zeta potential exceeding 30 mV and a PDI below 0.5. The stability was also maintained over a 14-d storage period. C-BT-NPs showed effective insecticidal activity against B. tabaci, with an LC50 of 1.51 g/L, outperforming thiamethoxam and offering efficient agricultural pest control. Furthermore, C-BT-NPs had minimal short-term impact on the growth of tomato plants, indicating that they are safety on plants. Therefore, the VOCs using nanoliposome preparation technology show promise in reducing reliance on conventional pesticides and present new approaches to managing agricultural pests.


Subject(s)
Hemiptera , Insecticides , Liposomes , Polycyclic Sesquiterpenes , Animals , Hemiptera/drug effects , Polycyclic Sesquiterpenes/pharmacology , Polycyclic Sesquiterpenes/chemistry , Insecticides/pharmacology , Insecticides/chemistry , Nanoparticles/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Solanum lycopersicum/parasitology , Volatile Organic Compounds/chemistry , Volatile Organic Compounds/pharmacology
20.
Dalton Trans ; 53(36): 15205-15214, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39221630

ABSTRACT

A novel lipoformulation was developed by encapsulating cationic (S^C)-cyclometallated gold(III) complex [Au(dppta)(N2Py-PZ-dtc)]+ (AuPyPZ) in liposomes. The liposomal form of compound AuPyPZ has a bactericidal action similar to that of the free drug without any appreciable effect on the viability of mammalian cells. Furthermore, the nanoformulation reduces metalloantibiotic-induced inhibition of hERG and the inhibition of cytochromes, significantly decreasing the potential liabilities of the metallodrug. The obtained metalloantibiotic liposomal formulation shows high stability and suitable properties for drug delivery, representing an effective strategy to fight against drug-resistant bacteria.


Subject(s)
Anti-Bacterial Agents , Gold , Liposomes , Microbial Sensitivity Tests , Liposomes/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Gold/chemistry , Gold/pharmacology , Humans , Drug Resistance, Bacterial/drug effects , Cell Survival/drug effects , Organogold Compounds/chemistry , Organogold Compounds/pharmacology , Organogold Compounds/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL