Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.856
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(11): e2307796121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437567

ABSTRACT

Cell-type-specific in vivo delivery of genome editing molecules is the next breakthrough that will drive biological discovery and transform the field of cell and gene therapy. Here, we discuss recent advances in the delivery of CRISPR-Cas genome editors either as preassembled ribonucleoproteins or encoded in mRNA. Both strategies avoid pitfalls of viral vector-mediated delivery and offer advantages including transient editor lifetime and potentially streamlined manufacturing capability that are already proving valuable for clinical use. We review current applications and future opportunities of these emerging delivery approaches that could make genome editing more efficacious and accessible in the future.


Subject(s)
Commerce , Gene Editing , Genetic Therapy , RNA, Messenger , Ribonucleoproteins
2.
Semin Immunol ; 61-64: 101664, 2022 11.
Article in English | MEDLINE | ID: mdl-36306664

ABSTRACT

Inflammatory bowel diseases (IBD) such as Crohn's Disease (CD) and Ulcerative Colitis (UC) are chronic, progressive, and relapsing disorders of the gastrointestinal tract (GIT), characterised by intestinal epithelial injury and inflammation. Current research shows that in addition to traditional anti-inflammatory therapy, resolution of inflammation and repair of the epithelial barrier are key biological requirements in combating IBD. Resolution mediators include endogenous lipids that are generated during inflammation, e.g., lipoxins, resolvins, protectins, maresins; and proteins such as Annexin A1 (ANXA1). Nanoparticles can specifically deliver these potent inflammation resolving mediators in a spatiotemporal manner to IBD lesions, effectively resolve inflammation, and promote a return to homoeostasis with minimal collateral damage. We discuss these exciting and timely concepts in this review.


Subject(s)
Annexin A1 , Inflammatory Bowel Diseases , Lipoxins , Humans , Annexin A1/metabolism , Inflammation/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammation Mediators/metabolism
3.
Proc Natl Acad Sci U S A ; 120(18): e2220036120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094132

ABSTRACT

SNIO-CBP, a single-nanometer iron oxide (SNIO) nanoparticle functionalized with a type I collagen-binding peptide (CBP), was developed as a T1-weighted MRI contrast agent with only endogenous elements for fast and noninvasive detection of liver fibrosis. SNIO-CBP exhibits 6.7-fold higher relaxivity compared to a molecular gadolinium-based collagen-binding contrast agent CM-101 on a per CBP basis at 4.7 T. Unlike most iron oxide nanoparticles, SNIO-CBP exhibits fast elimination from the bloodstream with a 5.7 min half-life, high renal clearance, and low, transient liver enhancement in healthy mice. We show that a dose of SNIO-CBP that is 2.5-fold lower than that for CM-101 has comparable imaging efficacy in rapid (within 15 min following intravenous injection) detection of hepatotoxin-induced liver fibrosis using T1-weighted MRI in a carbon tetrachloride-induced mouse liver injury model. We further demonstrate the applicability of SNIO-CBP in detecting liver fibrosis in choline-deficient L-amino acid-defined high-fat diet mouse model of nonalcoholic steatohepatitis. These results provide a platform with potential for the development of high relaxivity, gadolinium-free molecular MRI probes for characterizing chronic liver disease.


Subject(s)
Magnetite Nanoparticles , Nanoparticles , Mice , Animals , Contrast Media/chemistry , Liver Cirrhosis/pathology , Liver/pathology , Magnetic Resonance Imaging/methods , Disease Models, Animal , Magnetic Iron Oxide Nanoparticles , Collagen/analysis
4.
FASEB J ; 38(15): e23856, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39092913

ABSTRACT

Merozoites utilize sialic acids on the red blood cell (RBC) cell surface to rapidly adhere to and invade the RBCs. Newcastle disease virus (NDV) displays a strong affinity toward membrane-bound sialic acids. Incubation of NDV with the malaria parasites dose-dependently reduces its cellular viability. The antiplasmodial activity of NDV is specific, as incubation with Japanese encephalitis virus, duck enteritis virus, infectious bronchitis virus, and influenza virus did not affect the parasite propagation. Interestingly, NDV is reducing more than 80% invasion when RBCs are pretreated with the virus. Removal of the RBC surface proteins or the NDV coat proteins results in disruption of the virus binding to RBC. It suggests the involvement of specific protein: ligand interaction in virus binding. We established that the virus engages with the parasitized RBCs (PRBCs) through its hemagglutinin neuraminidase (HN) protein by recognizing sialic acid-containing glycoproteins on the cell surface. Blocking of the HN protein with free sialic acid or anti-HN antibodies abolished the virus binding as well as its ability to reduce parasite growth. Interestingly, the purified HN from the virus alone could inhibit the parasite's growth in a dose-dependent manner. NDV binds strongly to knobless murine parasite strain Plasmodium yoelii and restricted the parasite growth in mice. Furthermore, the virus was found to preferentially target the PRBCs compared to normal erythrocytes. Immunolocalization studies reveal that NDV is localized on the plasma membrane as well as weakly inside the PRBC. NDV causes neither any infection nor aggregation of the human RBCs. Our findings suggest that NDV is a potential candidate for developing targeted drug delivery platforms for the Plasmodium-infected RBCs.


Subject(s)
Erythrocytes , N-Acetylneuraminic Acid , Newcastle disease virus , Newcastle disease virus/physiology , Newcastle disease virus/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism , Animals , N-Acetylneuraminic Acid/metabolism , Humans , Plasmodium yoelii/metabolism , Mice , HN Protein/metabolism , Malaria/parasitology , Malaria/metabolism
5.
Mol Ther ; 32(8): 2762-2777, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38859589

ABSTRACT

This study demonstrates the potential of using biological nanoparticles to deliver RNA therapeutics targeting programmed death-ligand 1 (PD-L1) as a treatment strategy for cholangiocarcinoma (CCA). RNA therapeutics offer prospects for intracellular immune modulation, but effective clinical translation requires appropriate delivery strategies. Milk-derived nanovesicles were decorated with epithelial cellular adhesion molecule (EpCAM) aptamers and used to deliver PD-L1 small interfering RNA (siRNA) or Cas9 ribonucleoproteins directly to CCA cells. In vitro, nanovesicle treatments reduced PD-L1 expression in CCA cells while increasing degranulation, cytokine release, and tumor cell cytotoxicity when tumor cells were co-cultured with T cells or natural killer cells. Similarly, immunomodulation was observed in multicellular spheroids that mimicked the tumor microenvironment. Combining targeted therapeutic vesicles loaded with siRNA to PD-L1 with gemcitabine effectively reduced tumor burden in an immunocompetent mouse CCA model compared with controls. This proof-of-concept study demonstrates the potential of engineered targeted nanovesicle platforms for delivering therapeutic RNA cargoes to tumors, as well as their use in generating effective targeted immunomodulatory therapies for difficult-to-treat cancers such as CCA.


Subject(s)
B7-H1 Antigen , Cholangiocarcinoma , Immunotherapy , RNA, Small Interfering , Cholangiocarcinoma/therapy , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/immunology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Animals , Humans , Mice , Cell Line, Tumor , Immunotherapy/methods , RNA, Small Interfering/genetics , RNA, Small Interfering/administration & dosage , Nanoparticles/chemistry , Bile Duct Neoplasms/therapy , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/immunology , Tumor Microenvironment/immunology , Disease Models, Animal , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gemcitabine
6.
Semin Immunol ; 56: 101534, 2021 08.
Article in English | MEDLINE | ID: mdl-34836772

ABSTRACT

Lymph nodes are secondary lymphoid tissues in the body that facilitate the co-mingling of immune cells to enable and regulate the adaptive immune response. They are also tissues implicated in a variety of diseases, including but not limited to malignancy. The ability to access lymph nodes is thus attractive for a variety of therapeutic and diagnostic applications. As nanotechnologies are now well established for their potential in translational biomedical applications, their high relevance to applications that involve lymph nodes is highlighted. Herein, established paradigms of nanocarrier design to enable delivery to lymph nodes are discussed, considering the unique lymph node tissue structure as well as lymphatic system physiology. The influence of delivery mechanism on how nanocarrier systems distribute to different compartments and cells that reside within lymph nodes is also elaborated. Finally, current advanced nanoparticle technologies that have been developed to enable lymph node delivery are discussed.


Subject(s)
Drug Delivery Systems , Nanoparticles , Adaptive Immunity , Humans , Lymph Nodes , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Nanotechnology
7.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35173043

ABSTRACT

Safe and efficacious systemic delivery of messenger RNA (mRNA) to specific organs and cells in vivo remains the major challenge in the development of mRNA-based therapeutics. Targeting of systemically administered lipid nanoparticles (LNPs) coformulated with mRNA has largely been confined to the liver and spleen. Using a library screening approach, we identified that N-series LNPs (containing an amide bond in the tail) are capable of selectively delivering mRNA to the mouse lung, in contrast to our previous discovery that O-series LNPs (containing an ester bond in the tail) that tend to deliver mRNA to the liver. We analyzed the protein corona on the liver- and lung-targeted LNPs using liquid chromatography-mass spectrometry and identified a group of unique plasma proteins specifically absorbed onto the surface that may contribute to the targetability of these LNPs. Different pulmonary cell types can also be targeted by simply tuning the headgroup structure of N-series LNPs. Importantly, we demonstrate here the success of LNP-based RNA therapy in a preclinical model of lymphangioleiomyomatosis (LAM), a destructive lung disease caused by loss-of-function mutations in the Tsc2 gene. Our lung-targeting LNP exhibited highly efficient delivery of the mouse tuberous sclerosis complex 2 (Tsc2) mRNA for the restoration of TSC2 tumor suppressor in tumor and achieved remarkable therapeutic effect in reducing tumor burden. This research establishes mRNA LNPs as a promising therapeutic intervention for the treatment of LAM.


Subject(s)
Drug Delivery Systems/methods , Lymphangioleiomyomatosis/drug therapy , RNA, Messenger/administration & dosage , Animals , Female , Gene Transfer Techniques , Genetic Engineering/methods , Liposomes/chemistry , Liposomes/pharmacology , Lung/cytology , Lung/pathology , Lung Diseases/drug therapy , Lung Diseases/metabolism , Lymphangioleiomyomatosis/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Protein Corona/chemistry , Protein Corona/metabolism , RNA, Messenger/genetics , RNA, Messenger/pharmacology , RNA, Small Interfering/metabolism
8.
Nano Lett ; 24(27): 8351-8360, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38916238

ABSTRACT

Targeting telomere maintenance has emerged as a promising strategy for hepatocellular carcinoma (HCC) treatment. However, given the duality of the telomere-telomerase axis in telomere maintenance, a comprehensive strategy is urgently needed. Herein, we develop a poly(amino acid) (D-PAAs)-based strategy for spatiotemporal codelivery of telomerase inhibitor, BIBR1523, and AKT inhibitor, isobavachalcone. By leveraging D-PAAs' modifiability, we synthesize polymer-inhibitor conjugates (PB and PI) and a folic acid-decorated tumor-targeting vector (PF). These building blocks undergo micellization to fabricate a codelivery nanomedicine (P-BI@P-FA) by exploiting D-PAAs' noncovalent assembly. P-BI@P-FA improves the pharmacokinetics, tumor selectivity, and bioavailability of small molecule inhibitors and initiates a dual telomere-specific inhibition by combining telomerase deactivation with telomere disruption. Furthermore, a hybrid tumor-targeting magnetic nanosystem is designed using D-PAAs and manganese dioxide to showcase magnetic resonance imaging capacities. Our D-PAAs-based strategy addresses the pressing need for telomere-specific HCC treatment while allowing for diagnostic application, presenting a promising avenue for nanomedicine design.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Magnetic Resonance Imaging , Nanomedicine , Telomerase , Telomere , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Telomerase/antagonists & inhibitors , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/drug therapy , Humans , Nanomedicine/methods , Telomere/metabolism , Magnetic Resonance Imaging/methods , Animals , Mice , Cell Line, Tumor , Amino Acids/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use
9.
Nano Lett ; 24(17): 5154-5164, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38602357

ABSTRACT

Developing novel strategies for defeating osteoporosis has become a world-wide challenge with the aging of the population. In this work, novel supramolecular nanoagonists (NAs), constructed from alkaloids and phenolic acids, emerge as a carrier-free nanotherapy for efficacious osteoporosis treatment. These precision nanoagonists are formed through the self-assembly of berberine (BER) and chlorogenic acid (CGA), utilizing noncovalent electrostatic, π-π, and hydrophobic interactions. This assembly results in a 100% drug loading capacity and stable nanostructure. Furthermore, the resulting weights and proportions of CGA and BER within the NAs are meticulously controlled with strong consistency when the CGA/BER assembly feed ratio is altered from 1:1 to 1:4. As anticipated, our NAs themselves could passively target osteoporotic bone tissues following prolonged blood circulation, modulate Wnt signaling, regulate osteogenic differentiation, and ameliorate bone loss in ovariectomy-induced osteoporotic mice. We hope this work will open a new strategy to design efficient herbal-derived Wnt NAs for dealing with intractable osteoporosis.


Subject(s)
Berberine , Chlorogenic Acid , Osteoporosis , Osteoporosis/drug therapy , Animals , Mice , Berberine/pharmacology , Berberine/therapeutic use , Berberine/chemistry , Berberine/administration & dosage , Berberine/pharmacokinetics , Chlorogenic Acid/chemistry , Chlorogenic Acid/pharmacology , Chlorogenic Acid/therapeutic use , Chlorogenic Acid/administration & dosage , Female , Humans , Osteogenesis/drug effects , Bone and Bones/drug effects , Bone and Bones/pathology , Nanostructures/chemistry , Nanostructures/therapeutic use
10.
Biochem Biophys Res Commun ; 725: 150260, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38878760

ABSTRACT

This study introduces an innovative brain-targeted drug delivery system, RVG-Exo/CBD, utilizing rabies virus glycoprotein (RVG)-engineered exosomes for encapsulating cannabidiol (CBD). The novel delivery system was meticulously characterized, confirming the maintenance of exosomal integrity, size, and successful drug encapsulation with a high drug loading rate of 83.0 %. Evaluation of the RVG-Exo/CBD's brain-targeting capability demonstrated superior distribution and retention in brain tissue compared to unmodified exosomes, primarily validated through in vivo fluorescence imaging. The efficacy of this delivery system was assessed using a behavioral sensitization model in mice, where RVG-Exo/CBD notably suppressed methamphetamine-induced hyperactivity more effectively than CBD alone, indicating a reduction in effective dose and enhanced bioavailability. Overall, the RVG-Exo/CBD system emerges as a promising strategy for enhancing the therapeutic efficacy and safety of CBD, particularly for neurological applications, highlighting its potential for addressing the limitations associated with traditional CBD administration in clinical settings.


Subject(s)
Brain , Cannabidiol , Cannabidiol/administration & dosage , Cannabidiol/chemistry , Cannabidiol/pharmacology , Animals , Brain/metabolism , Brain/drug effects , Mice , Male , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycoproteins/administration & dosage , Drug Delivery Systems/methods , Peptide Fragments , Viral Proteins
11.
Small ; 20(20): e2306909, 2024 May.
Article in English | MEDLINE | ID: mdl-38100246

ABSTRACT

Helicobacter pylori (H. pylori) infection presents increasing challenges to antibiotic therapies in limited penetration through gastric mucus, multi-drug resistance (MDR), biofilm formation, and intestinal microflora dysbiosis. To address these problems, herein, a mucus-penetrating phototherapeutic nanomedicine (RLs@T780TG) against MDR H. pylori infection is engineered. The RLs@T780TG is assembled with a near-infrared photosensitizer T780T-Gu and an anionic component rhamnolipids (RLs) for deep mucus penetration and light-induced anti-H. pylori performances. With optimized suitable size, hydrophilicity and weak negative surface, the RLs@T780TG can effectively penetrate through the gastric mucus layer and target the inflammatory site. Subsequently, under irradiation, the structure of RLs@T780TG is disrupted and facilitates the T780T-Gu releasing to target the H. pylori surface and ablate multi-drug resistant (MDR) H. pylori. In vivo, RLs@T780TG phototherapy exhibits impressive eradication against H. pylori. The gastric lesions are significantly alleviated and intestinal bacteria balance is less affected than antibiotic treatment. Summarily, this work provides a potential nanomedicine design to facilitate in vivo phototherapy in treatment of H. pylori infection.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Mucus , Helicobacter pylori/drug effects , Helicobacter Infections/drug therapy , Mucus/metabolism , Animals , Phototherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Drug Resistance, Multiple, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Glycolipids/chemistry , Glycolipids/pharmacology , Mice , Administration, Oral
12.
Small ; 20(21): e2306482, 2024 May.
Article in English | MEDLINE | ID: mdl-38109123

ABSTRACT

Inflammatory bowel disease (IBD) has become a globally prevalent chronic disease with no causal therapeutic options. Targeted drug delivery systems with selectivity for inflamed areas in the gastrointestinal tract promise to reduce severe drug-related side effects. By creating three distinct nanostructures (vesicles, spherical, and wormlike micelles) from the same amphiphilic block copolymer poly(butyl acrylate)-block-poly(ethylene oxide) (PBA-b-PEO), the effect of nanoparticle shape on human mucosal penetration is systematically identified. An Ussing chamber technique is established to perform the ex vivo experiments on human colonic biopsies, demonstrating that the shape of polymeric nanostructures represents a rarely addressed key to tissue selectivity required for efficient IBD treatment. Wormlike micelles specifically enter inflamed mucosa from patients with IBD, but no significant uptake is observed in healthy tissue. Spheres (≈25 nm) and vesicles (≈120 nm) enter either both normal and inflamed tissue types or do not penetrate any tissue. According to quantitative image analysis, the wormlike nanoparticles localize mainly within immune cells, facilitating specific targeting, which is crucial for further increasing the efficacy of IBD treatment. These findings therefore demonstrate the untapped potential of wormlike nanoparticles not only to selectively target the inflamed human mucosa, but also to target key pro-inflammatory cells.


Subject(s)
Inflammatory Bowel Diseases , Micelles , Polymers , Humans , Polymers/chemistry , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Nanoparticles/chemistry , Inflammation/drug therapy , Inflammation/pathology , Drug Delivery Systems
13.
Small ; : e2402792, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940386

ABSTRACT

Adjuvants play a critical role in the induction of effective immune responses by vaccines. Here, a self-assembling nanovaccine platform that integrates adjuvant functions into the delivery vehicle is prepared. Cationic Lentinan (CLNT) is mixed with ovalbumin (OVA) to obtain a self-assembling nanovaccine (CLNTO nanovaccine), which induces the uptake and maturation of bone marrow dendritic cells (BMDCs) via the toll-like receptors 2/4 (TLR2/4) to produce effective antigen cross-presentation. CLNTO nanovaccines target lymph nodes (LNs) and induce a robust OVA-specific immune response via TLR and tumor necrosis factor (TNF) signaling pathways, retinoic acid-inducible gene I (RIG-I) receptor, and cytokine-cytokine receptor interactions. In addition, CLNTO nanovaccines are found that promote the activation of follicular helper T (Tfh) cells and induce the differentiation of germinal center (GC) B cells into memory B cells and plasma cells, thereby enhancing the immune response. Vaccination with CLNTO nanovaccine significantly inhibits the growth of ovalbumin (OVA)-expressing B16 melanoma cell (B16-OVA) tumors, indicating its great potential for cancer immunotherapy. Therefore, this study presents a simple, safe, and effective self-assembling nanovaccine that induces helper T cell 1 (Th1) and helper T cell (Th2) immune responses, making it an effective vaccine delivery system.

14.
Chembiochem ; 25(8): e202300831, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38408302

ABSTRACT

Peptide and protein nanostructures with tunable structural features, multifunctionality, biocompatibility and biomolecular recognition capacity enable development of efficient targeted drug delivery tools for precision medicine applications. In this review article, we present various techniques employed for the synthesis and self-assembly of peptides and proteins into nanostructures. We discuss design strategies utilized to enhance their stability, drug-loading capacity, and controlled release properties, in addition to the mechanisms by which peptide nanostructures interact with target cells, including receptor-mediated endocytosis and cell-penetrating capabilities. We also explore the potential of peptide and protein nanostructures for precision medicine, focusing on applications in personalized therapies and disease-specific targeting for diagnostics and therapeutics in diseases such as cancer.


Subject(s)
Nanostructures , Precision Medicine , Drug Delivery Systems/methods , Peptides/chemistry , Nanostructures/therapeutic use , Nanostructures/chemistry , Pharmaceutical Preparations
15.
Annu Rev Biomed Eng ; 25: 233-256, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37000965

ABSTRACT

Accompanying the increasing translational impact of immunotherapeutic strategies to treat and prevent disease has been a broadening interest across both bioscience and bioengineering in the lymphatic system. Herein, the lymphatic system physiology, ranging from its tissue structures to immune functions and effects, is described. Design principles and engineering approaches to analyze and manipulate this tissue system in nanoparticle-based drug delivery applications are also elaborated.


Subject(s)
Bioengineering , Drug Delivery Systems , Humans , Nanotechnology , Lymphatic System
16.
Neurochem Res ; 49(9): 2364-2378, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38837091

ABSTRACT

As an element of the cellular signaling systems, extracellular vesicles (EVs) exhibit many desirable traits for usage as targeted delivery vehicles. When administered, EVs cause little to no toxic or immune response, stay in circulation for longer periods compared to synthetic carriers, preferentially accumulate in tissues that are the same or similar to their cell-of-origin and can pass through the blood-brain barrier. Combined, these traits make neural EVs a particularly promising tool for delivering drugs to the brain. This study aims to combine tissue and EVs engineering to prepare neural differentiated cells derived EVs that exhibit neural properties, to develop an effective, tissue-homing drug and gene delivery platform for the brain. Early neural differentiated cell-derived EVs were produced with neural characteristics from neural differentiated human neonatal dermal fibroblasts. The EVs carried key neural proteins such as Nestin, Sox2 and Doublecortin. The cellular uptake of early neural differentiated cell-derived EVs was higher compared to non-neural EVs during in vitro uptake assays on neuroblastoma cells. Moreover, eND-EVs were significantly decreased the viability of neuroblastoma cells. In conclusion, this study revealed that early neural differentiated cell-derived EVs have potential as a promising drug carrier for the treatment of various neural disorders.


Subject(s)
Extracellular Vesicles , Neural Stem Cells , Neuroblastoma , Extracellular Vesicles/metabolism , Humans , Neural Stem Cells/metabolism , Cell Line, Tumor , Drug Delivery Systems/methods , Cell Differentiation/physiology , Cell Survival/physiology , Cell Survival/drug effects , Fibroblasts/metabolism , SOXB1 Transcription Factors
17.
Mol Pharm ; 21(3): 1077-1089, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38346386

ABSTRACT

Folic acid (FA) has been widely engineered to promote the targeted delivery of FA-modified nanoparticles (NPs) by recognizing the folate receptor α (FRα). However, the efficacy of FA-targeted therapy significantly varied with the abundance of FRα and natural immunoglobulin levels in different tumors. Therefore, a sequential therapy of dexamethasone (Dex)-induced FRα amplification and immunosuppression combined with FA-functionalized doxorubicin (DOX) micelles to synergistically suppress tumor proliferation was proposed in this study. In brief, a pH/reduction-responsive FA-functionalized micelle (FCSD) was obtained by grafting FA, derivatization-modified cholesterol, and 2,3-dimethylmaleic anhydride onto a chitosan oligosaccharide. The obtained FCSD/DOX NPs can effectively deliver DOX in tumors, and their targeting efficiency can be further improved with Dex pretreatment to decrease the immunoglobulin M (IgM) content in serum and amplify FRα levels on the surface of M109 cells. After internalization, charge reversal and disulfide bond breakage of FCSD vectors under the stimulation of tumor extracellular pH (pHe) and intracellular glutathione (GSH) would contribute to the disintegration of vectors and the rapid release of DOX. The sequential therapy that combined Dex pretreatment and targeted chemotherapy by FCSD/DOX NPs demonstrated superior tumor suppression compared with monotherapy, which is expected to provide a potential strategy for FRα-positive lung cancer patients.


Subject(s)
Lung Neoplasms , Nanoparticles , Humans , Drug Carriers/chemistry , Lung Neoplasms/drug therapy , Folic Acid/chemistry , Doxorubicin , Micelles , Nanoparticles/chemistry , Dexamethasone , Drug Delivery Systems , Hydrogen-Ion Concentration
18.
Nitric Oxide ; 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39243805

ABSTRACT

Hydrogen sulfide (H2S) has emerged as a potent therapeutic agent with diverse physiological functions, including vasodilation, anti-inflammation, and cytoprotection. However, its clinical application is limited due to its volatility and potential toxicity at high concentrations. To address these challenges, researchers have developed various H2S prodrugs that release H2S in a controlled and targeted manner. The review underscores the importance of targeting and delivery strategies in maximizing the therapeutic potential of H2S, a gasotransmitter with diverse physiological functions and therapeutic effects. By summarizing recent advancements, the review provides valuable insights for researchers and clinicians interested in harnessing the therapeutic benefits of H2S while minimizing off-target effects and toxicity. The integration of novel targeting and delivery approaches not only enhances the efficacy of H2S-based therapeutics but also expands the scope of potential applications, offering promising avenues for the development of new treatments for a variety of diseases and disorders.

19.
Pharmacol Res ; 199: 107022, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043691

ABSTRACT

Macrophages, as highly phenotypic plastic immune cells, play diverse roles in different pathological conditions. Changing and controlling the phenotypes of macrophages is considered a novel potential therapeutic intervention. Meanwhile, specific transmembrane proteins anchoring on the surface of the macrophage membrane are relatively conserved, supporting its functional properties, such as inflammatory chemotaxis and tumor targeting. Thus, a series of drug delivery systems related to specific macrophage membrane proteins are commonly used to treat chronic inflammatory diseases. This review summarizes macrophages-based strategies for chronic diseases, discusses the regulation of macrophage phenotypes and their polarization processes, and presents how to design and apply the site-specific targeted drug delivery systems in vivo based on the macrophages and their derived membrane receptors. It aims to provide a better understanding of macrophages in immunoregulation and proposes macrophages-based targeted therapeutic approaches for chronic diseases.


Subject(s)
Drug Delivery Systems , Neoplasms , Humans , Phenotype , Macrophages/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Chronic Disease
20.
Pharm Res ; 41(2): 263-279, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38263341

ABSTRACT

INTRODUCTION: Exosomes are extracellular vesicles in the range of 40-150 nm released from the cell membrane. Exosomes secreted by keratinocytes can communicate with other keratinocytes and immune cells with specific biomarkers at their surface, which may be effective on inflammation of psoriasis and its pathogenesis. OBJECTIVE: The present study aimed to formulate and study effectiveness of an exosomal delivery system of tofacitinib (TFC). METHODS: TFC was loaded by different methods in exosomes and then characterized for particle size, zeta potential, drug loading efficiency, and release efficiency. By comparing these parameters, the probe sonication method was chosen to load TFC into exosomes. The MTT assay was used to compare the cytotoxicity of the free drug with the TFC-loaded exosomes (TFC-Exo), and Real-time PCR was used to determine the expression levels of several genes involved in psoriasis expressed in the A-431 keratinocyte and their suppression after treatment. Animal model of psoriasis was induced in BALB/c mice by imiquimod and the efficacy of free TFC, and TFC-Exo were studies on macroscopic appearance and histopathological symptoms. RESULTS: Exosomes encapsulating TFC showed lower cytotoxicity in MTT assay, higher suppression the expression of TNF-a, IL-23, IL-6, and IL-15 genes in real-time PCR and better therapeutic effect on animal models compered to free TFC. CONCLUSIONS: This method of drug delivery for TFC may be effective on enhancing its therapeutic effects and reduction its side effects favorably in chronic administration.


Subject(s)
Exosomes , Piperidines , Psoriasis , Pyrimidines , Animals , Mice , Exosomes/metabolism , Keratinocytes/metabolism , Psoriasis/drug therapy , Models, Animal , Disease Models, Animal , Mice, Inbred BALB C , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL