Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 564
Filtrar
1.
Mol Ther ; 32(8): 2762-2777, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38859589

RESUMEN

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.


Asunto(s)
Antígeno B7-H1 , Colangiocarcinoma , Inmunoterapia , ARN Interferente Pequeño , Colangiocarcinoma/terapia , Colangiocarcinoma/metabolismo , Colangiocarcinoma/inmunología , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Animales , Humanos , Ratones , Línea Celular Tumoral , Inmunoterapia/métodos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/administración & dosificación , Nanopartículas/química , Neoplasias de los Conductos Biliares/terapia , Neoplasias de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/inmunología , Microambiente Tumoral/inmunología , Modelos Animales de Enfermedad , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Gemcitabina
2.
Nano Lett ; 24(5): 1570-1578, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38287297

RESUMEN

Glioblastoma (GBM) is the most complex and lethal primary brain cancer. Adequate drug diffusion and penetration are essential for treating GBM, but how the spatial heterogeneity in GBM impacts drug diffusion and transport is poorly understood. Herein, we report a new method, photoactivation of plasmonic nanovesicles (PANO), to measure molecular diffusion in the extracellular space of GBM. By examining three genetically engineered GBM mouse models that recapitulate key clinical features including the angiogenic core and diffuse infiltration, we found that the tumor margin has the lowest diffusion coefficient (highest tortuosity) compared with the tumor core and surrounding brain tissue. Analysis of the cellular composition shows that tortuosity in the GBM is strongly correlated with neuronal loss and astrocyte activation. Our all-optical measurement reveals the heterogeneous GBM microenvironment and highlights the tumor margin as a diffusion barrier for drug transport in the brain, with implications for therapeutic delivery.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Ratones , Animales , Glioblastoma/patología , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/patología , Línea Celular Tumoral , Espacio Extracelular , Microambiente Tumoral
3.
Nano Lett ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38856668

RESUMEN

Cell membrane-based nanovesicles (CMNVs) play pivotal roles in biomolecular transportation in living organisms and appear as attractive bioinformed nanomaterials for theranostic applications. However, the current surface-engineering technologies are limited in flexibility and orthogonality, making it challenging to simultaneously display multiple different ligands on the CMNV surface in a precisely controlled manner. Here, we developed a DNA scaffold-programmed approach to orthogonally engineer CMNVs with versatile ligands. The designed DNA scaffolds can rapidly anchor onto the CMNV surface, and their unique sequences and hybridized properties enable independent control of the loading of multiple different types of biomolecules on the CMNVs. As a result, the orthogonal engineering of CMNVs with a renal targeted peptide and a therapeutic protein at controlled ratios demonstrated an enhanced renal targeting and repair potential in vivo. This study highlights that a DNA scaffold-programmed platform can provide a potent means for orthogonal and flexible surface engineering of CMNVs for diverse therapeutic purposes.

4.
Semin Cancer Biol ; 90: 73-100, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36773820

RESUMEN

Extracellular vesicles (EVs) function as a mode of intercellular communication and molecular transfer to elicit diverse biological/functional response. Accumulating evidence has highlighted that EVs from immune, tumour, stromal cells and even bacteria and parasites mediate the communication of various immune cell types to dynamically regulate host immune response. EVs have an innate capacity to evade recognition, transport and transfer functional components to target cells, with subsequent removal by the immune system, where the immunological activities of EVs impact immunoregulation including modulation of antigen presentation and cross-dressing, immune activation, immune suppression, and immune surveillance, impacting the tumour immune microenvironment. In this review, we outline the recent progress of EVs in immunorecognition and therapeutic intervention in cancer, including vaccine and targeted drug delivery and summarise their utility towards clinical translation. We highlight the strategies where EVs (natural and engineered) are being employed as a therapeutic approach for immunogenicity, tumoricidal function, and vaccine development, termed immuno-EVs. With seminal studies providing significant progress in the sequential development of engineered EVs as therapeutic anti-tumour platforms, we now require direct assessment to tune and improve the efficacy of resulting immune responses - essential in their translation into the clinic. We believe such a review could strengthen our understanding of the progress in EV immunobiology and facilitate advances in engineering EVs for the development of novel EV-based immunotherapeutics as a platform for cancer treatment.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Humanos , Vesículas Extracelulares/metabolismo , Neoplasias/patología , Presentación de Antígeno , Vigilancia Inmunológica , Inmunoterapia , Microambiente Tumoral
5.
Proteomics ; 24(11): e2200145, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38214697

RESUMEN

The ability of trophectodermal cells (outer layer of the embryo) to attach to the endometrial cells and subsequently invade the underlying matrix are critical stages of embryo implantation during successful pregnancy establishment. Extracellular vesicles (EVs) have been implicated in embryo-maternal crosstalk, capable of reprogramming endometrial cells towards a pro-implantation signature and phenotype. However, challenges associated with EV yield and direct loading of biomolecules limit their therapeutic potential. We have previously established generation of cell-derived nanovesicles (NVs) from human trophectodermal cells (hTSCs) and their capacity to reprogram endometrial cells to enhance adhesion and blastocyst outgrowth. Here, we employed a rapid NV loading strategy to encapsulate potent implantation molecules such as HB-EGF (NVHBEGF). We show these loaded NVs elicit EGFR-mediated effects in recipient endometrial cells, activating kinase phosphorylation sites that modulate their activity (AKT S124/129, MAPK1 T185/Y187), and downstream signalling pathways and processes (AKT signal transduction, GTPase activity). Importantly, they enhanced target cell attachment and invasion. The phosphoproteomics and proteomics approach highlight NVHBEGF-mediated short-term signalling patterns and long-term reprogramming capabilities on endometrial cells which functionally enhance trophectodermal-endometrial interactions. This proof-of-concept study demonstrates feasibility in enhancing the functional potency of NVs in the context of embryo implantation.


Asunto(s)
Vesículas Extracelulares , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Femenino , Vesículas Extracelulares/metabolismo , Endometrio/metabolismo , Endometrio/citología , Esferoides Celulares/metabolismo , Esferoides Celulares/citología , Trofoblastos/metabolismo , Trofoblastos/citología , Implantación del Embrión , Adhesión Celular , Transducción de Señal , Proteómica/métodos , Embarazo
6.
Curr Atheroscler Rep ; 26(7): 289-304, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38805145

RESUMEN

PURPOSE OF REVIEW: In this review, we explore the intriguing and evolving connections between bacterial extracellular membrane nanovesicles (BEMNs) and atherosclerosis development, highlighting the evidence on molecular mechanisms by which BEMNs can promote the athero-inflammatory process that is central to the progression of atherosclerosis. RECENT FINDINGS: Atherosclerosis is a chronic inflammatory disease primarily driven by metabolic and lifestyle factors; however, some studies have suggested that bacterial infections may contribute to the development of both atherogenesis and inflammation in atherosclerotic lesions. In particular, the participation of BEMNs in atherosclerosis pathogenesis has attracted special attention. We provide some general insights into how the immune system responds to potential threats such as BEMNs during the development of atherosclerosis. A comprehensive understanding of contribution of BEMNs to atherosclerosis pathogenesis may lead to the development of targeted interventions for the prevention and treatment of the disease.


Asunto(s)
Aterosclerosis , Vesículas Extracelulares , Aterosclerosis/microbiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Humanos , Vesículas Extracelulares/metabolismo , Animales , Inflamación/metabolismo , Bacterias/metabolismo , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/metabolismo
7.
Cell Commun Signal ; 22(1): 178, 2024 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-38475787

RESUMEN

BACKGROUND: Carthamus tinctorius L., a traditional herbal medicine used for atherosclerosis (AS), lacks a clear understanding of its therapeutic mechanisms. This study aimed to investigate the therapeutic effects and mechanisms of Carthamus tinctorius L.-derived nanovesicles (CDNVs) in AS treatment. METHODS: CDNVs were isolated and characterized using improved isolation methods. Transmission electron microscopy, nanoparticle tracking analysis, and protein analysis confirmed their morphology, size, and protein composition. Small RNA sequencing was performed to identify the miRNA profile of CDNVs, and bioinformatics analysis was used to determine their potential biological roles. In vivo biodistribution and toxicity studies were conducted in mice to assess the stability and safety of orally administered CDNVs. The anti-atherosclerotic effects of CDNVs were evaluated in ApoE-/- mice through plaque burden analysis. The protective effects of CDNVs on ox-LDL-treated endothelial cells were assessed through proliferation, apoptosis, reactive oxygen species activation, and monocyte adhesion assays. miRNA and mRNA sequencing of CDNV-treated endothelial cells were performed to explore their regulatory effects and potential target genes. RESULTS: CDNVs were successfully isolated and purified from Carthamus tinctorius L. tissue lysates. They exhibited a saucer-shaped or cup-shaped morphology, with an average particle size of 142.6 ± 0.7 nm, and expressed EV markers CD63 and TSG101. CDNVs contained proteins, small RNAs, and metabolites, including the therapeutic compound HSYA. Small RNA sequencing identified 95 miRNAs, with 10 common miRNAs accounting for 72.63% of the total miRNAs. These miRNAs targeted genes involved in cell adhesion, apoptosis, and cell proliferation, suggesting their relevance in cardiovascular disease. Orally administered CDNVs were stable in the gastrointestinal tract, absorbed into the bloodstream, and accumulated in the liver, lungs, heart, and aorta. They significantly reduced the burden of atherosclerotic plaques in ApoE-/- mice and exhibited superior effects compared to HSYA. In vitro studies demonstrated that CDNVs were taken up by HUVECs, promoted proliferation, attenuated ox-LDL-induced apoptosis and ROS activation, and reduced monocyte adhesion. CDNV treatment resulted in significant changes in miRNA and mRNA expression profiles of HUVECs, with enrichment in inflammation-related genes. CXCL12 was identified as a potential direct target of miR166a-3p. CONCLUSION: CDNVs isolated from Carthamus tinctorius L. tissue lysates represent a promising oral therapeutic option for cardiovascular diseases. The delivery of miRNAs by CDNVs regulates inflammation-related genes, including CXCL12, in HUVECs, suggesting their potential role in modulating endothelial inflammation. These findings provide valuable insights into the therapeutic potential of CDNVs and their miRNAs in cardiovascular disease.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Carthamus tinctorius , MicroARNs , Ratones , Animales , Células Endoteliales/metabolismo , Carthamus tinctorius/genética , Carthamus tinctorius/metabolismo , Enfermedades Cardiovasculares/metabolismo , Distribución Tisular , Ratones Noqueados para ApoE , MicroARNs/genética , Aterosclerosis/metabolismo , Inflamación/metabolismo , Apoptosis , ARN Mensajero/metabolismo , Apolipoproteínas E/metabolismo
8.
Pharmacol Res ; 204: 107202, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38704110

RESUMEN

Plant-derived extracellular vesicles (PDEV) constitute nanoscale entities comprising lipids, proteins, nucleic acids and various components enveloped by the lipid bilayers of plant cells. These vesicles play a crucial role in facilitating substance and information transfer not only between plant cells but also across different species. Owing to its safety, stability, and the abundance of raw materials, this substance has found extensive utilization in recent years within research endeavors aimed at treating various diseases. This article provides an overview of the pathways and biological characteristics of PDEV, along with the prevalent methods employed for its isolation, purification, and storage. Furthermore, we comprehensively outline the therapeutic implications of diverse sources of PDEV in musculoskeletal system disorders. Additionally, we explore the utilization of PDEV as platforms for engineering drug carriers, aiming to delve deeper into the significance and potential contributions of PDEV in the realm of the musculoskeletal system.


Asunto(s)
Vesículas Extracelulares , Músculo Esquelético , Plantas , Animales , Humanos , Portadores de Fármacos/química , Vesículas Extracelulares/metabolismo , Músculo Esquelético/metabolismo , Plantas/metabolismo
9.
Mol Biol Rep ; 51(1): 425, 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38492036

RESUMEN

Small extracellular vesicles (sEVs) isolated from animal sources are among the most investigated types of cell-free therapeutic tools to cure different diseases. sEVs have been isolated from a variety of sources, ranging from prokaryotes to animals and plants. Human-derived sEVs have many uses in pre- and clinical studies in medicine and drug delivery, while plant-derived EVs, also known as plant-derived nanovesicles (PDNVs), have not been widely investigated until the second decade of the 21st century. For the past five years, there has been a rapid rise in the use of plant EVs as a therapeutic tool due to the ease of massive production with high efficacy and yield of preparation. Plant EVs contain various active biomolecules such as proteins, regulatory RNAs, and secondary metabolites and play a key role in inter-kingdom communications. Many studies have already investigated the potential application of plant EVs in preventing and treating cancer, inflammation, infectious diseases, and tissue regeneration with no sign of toxicity and are therefore considered safe. However, due to a lack of universal markers, the properties of plant EVs have not been extensively studied. Concerns regarding the safety and therapeutic function of plant EVs derived from genetically modified plants have been raised. In this paper, we review the physiological role of EVs in plants. Moreover, we focus on molecular and cellular mechanisms involved in the therapeutic effects of plant EVs on various human diseases. We also provide detailed information on the methodological aspects of plant EV isolation and analysis, which could pave the way for future clinical translation.


Asunto(s)
Vesículas Extracelulares , Animales , Humanos , Sistemas de Liberación de Medicamentos , Inflamación , ARN
10.
J Biochem Mol Toxicol ; 38(1): e23540, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37728183

RESUMEN

Dose-dependent heart failure is a major complication of the clinical use of doxorubicin (Dox), one of the most potent chemotherapeutic agents. Effective adjuvant therapy is required to prevent Dox-induced cardiotoxicity. Currently, plant-derived exosome-like nanovesicle (PELNV) has revealed their salubrious antioxidant and immunological regulating actions in various disease models. In this study, we isolated, purified and characterized Beta vulgaris-derived exosome-like nanovesicle (BELNV). Dox or normal saline was given to HL-1 cells (3 µM) and 8-week C57BL/6N mice (5 mg/kg bodyweight per week for 4 weeks) to establish the in vitro and in vivo model of Dox-induced cardiotoxicity. Administration of BELNV significantly alleviated chronic Dox-induced cardiotoxicity in terms of echocardiographic and histological results. A reduced malondialdehyde (MDA), increased ratio of glutathione (GSH) to oxidized glutathione (GSSG) and levels of system xc- and glutathione peroxidase 4 were observed, indicating that DOX-stimulated ferroptosis was reversed by BELNV. Besides, the safety of BELNV was also validated since no liver, spleen, and kidney toxicity induced by BELNV was observed. These findings provide evidence that BELNV may act as a novel therapeutic biomaterial for patients undergoing adverse effects of Dox, at least partly mediated by inhibiting Dox-induced ferroptosis.


Asunto(s)
Beta vulgaris , Exosomas , Ferroptosis , Humanos , Ratones , Animales , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Miocardio/metabolismo , Beta vulgaris/metabolismo , Exosomas/metabolismo , Ratones Endogámicos C57BL , Doxorrubicina/efectos adversos , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Disulfuro de Glutatión/farmacología , Disulfuro de Glutatión/uso terapéutico , Estrés Oxidativo , Miocitos Cardíacos/metabolismo
11.
J Clin Periodontol ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951121

RESUMEN

AIM: To identify an optimized strategy for the large-scale production of nanovesicles (NVs) that preserve the biological properties of exosomes (EXOs) for use in periodontal regeneration. MATERIALS AND METHODS: NVs from dental follicle stem cells (DFSCs) were prepared through extrusion, and EXOs from DFSCs were isolated. The yield of both extruded NVs (eNVs) and EXOs were quantified through protein concentration and particle number analyses. Their pro-migration, pro-proliferation and pro-osteogenesis capacities were compared subsequently in vitro. Additionally, proteomics analysis was conducted. To further evaluate the periodontal regeneration potential of eNVs and EXOs, they were incorporated into collagen sponges and transplanted into periodontal defects in rats. In vivo imaging and H&E staining were utilized to verify their biodistribution and safety. Micro-Computed Tomography analysis and histological staining were performed to examine the regeneration of periodontal tissues. RESULTS: The yield of eNVs was nearly 40 times higher than that of EXOs. Interestingly, in vitro experiments indicated that the pro-migration and pro-proliferation abilities of eNVs were superior, and the pro-osteogenesis potential was comparable to EXOs. More importantly, eNVs exhibited periodontal regenerative potential similar to that of EXOs. CONCLUSIONS: Extrusion has proven to be an efficient method for generating numerous eNVs with the potential to replace EXOs in periodontal regeneration.

12.
Mol Ther ; 31(5): 1207-1224, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-36245129

RESUMEN

Developing strategies toward safe and effective drug delivery into the central nervous system (CNS) with improved targeting abilities and reduced off-target effects is crucial. CNS-targeted drug carriers made of synthetic molecules raise concerns about their biodegradation, clearance, immune responses, and neurotoxicity. Cell-derived nanovesicles (CDNs) have recently been applied in CNS-targeted drug delivery, because of their intrinsic stability, biocompatibility, inherent homing capability, and the ability to penetrate through biological barriers, including the blood-brain barrier. Among these CDNs, extracellular vesicles and exosomes are the most studied because their surface can be engineered and modified to cater to brain targeting. In this review, we focus on the application of CDNs in brain-targeted drug delivery to treat neurological diseases. We cover recently developed methods of exosome derivation and engineering, including exosome-like particles, hybrid exosomes, exosome-associated adeno-associated viruses, and envelope protein nanocages. Finally, we discuss the limitations and project the future development of the CDN-based brain-targeted delivery systems, and conclude that engineered CDNs hold great potential in the treatment of neurological diseases.


Asunto(s)
Exosomas , Vesículas Extracelulares , Sistemas de Liberación de Medicamentos/métodos , Encéfalo , Exosomas/metabolismo , Barrera Hematoencefálica/metabolismo
13.
J Nanobiotechnology ; 22(1): 269, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764018

RESUMEN

Symbiotic microbial communities are crucial for human health, and dysbiosis is associated with various diseases. Plant-derived nanovesicles (PDNVs) have a lipid bilayer structure and contain lipids, metabolites, proteins, and RNA. They offer unique advantages in regulating microbial community homeostasis and treating diseases related to dysbiosis compared to traditional drugs. On the one hand, lipids on PDNVs serve as the primary substances that mediate specific recognition and uptake by bacteria. On the other hand, due to the multifactorial nature of PDNVs, they have the potential to enhance growth and survival of beneficial bacterial while simultaneously reducing the pathogenicity of harmful bacteria. In addition, PDNVs have the capacity to modulate bacterial metabolism, thus facilitating the establishment of a harmonious microbial equilibrium and promoting stability within the microbiota. These remarkable attributes make PDNVs a promising therapeutic approach for various conditions, including periodontitis, inflammatory bowel disease, and skin infection diseases. However, challenges such as consistency, isolation methods, and storage need to be addressed before clinical application. This review aims to explore the value of PDNVs in regulating microbial community homeostasis and provide recommendations for their use as novel therapeutic agents for health protection.


Asunto(s)
Microbiota , Humanos , Plantas , Bacterias/metabolismo , Disbiosis/microbiología , Animales , Nanopartículas/química , Nanoestructuras/química , Periodontitis/microbiología
14.
J Nanobiotechnology ; 22(1): 607, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39379937

RESUMEN

Ulcerative colitis (UC) belongs to chronic inflammatory disease with a relapsing characterization. Conventional oral drugs of UC are restricted in clinical by premature degradation in the gastrointestinal tract, modest efficacy, and adverse effects. CX5461 can treat autoimmune disease, immunological rejection, and vascular inflammation. However, low solubility, intravenous administration, and non-inflammatory targeting limited its clinical application. Herein, this work aims to develop Sophora Flavescens-derived exosomes-like nanovesicles carrying CX5461 (SFELNVs@CX5461) for efficient CX5461 oral delivery for UC therapy. We identified SFELNVs as nano-diameter (80 nm) with negative zeta potential (-32mV). Cellular uptake has shown that SFELNVs were targeted uptake by macrophages, thus increasing drug concentration. Additionally, oral SFELNVs@CX5461 exhibited good safety and stability, as well as inflammation-targeting ability in the gastrointestinal tract of dextran sodium sulfate (DSS)-induced colitis mice. In vivo, oral administration of SFELNVs and CX5461 could relieve mice colitis. More importantly, combined SFELNVs and CX5461 alleviated mice colitis by inhibiting pro-inflammatory factors (TNF-α, IL-1ß, and IL-6) expression and promoting M2 macrophage polarization. Furthermore, SFELNVs promoted M2 polarization by miR4371c using miRNA sequencing. Our results suggest that SFELNVs@CX5461 represents a novel orally therapeutic drug that can ameliorate colitis, and a promising targeting strategy for safe UC therapy.


Asunto(s)
Colitis , Sulfato de Dextran , Exosomas , Sophora , Animales , Ratones , Exosomas/metabolismo , Administración Oral , Sophora/química , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Masculino , Células RAW 264.7 , Ratones Endogámicos C57BL , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Nanopartículas/química , Humanos , Sophora flavescens
15.
J Nanobiotechnology ; 22(1): 190, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637808

RESUMEN

Acute lung injury (ALI) is generally caused by severe respiratory infection and characterized by overexuberant inflammatory responses and inefficient pathogens-containing, the two major processes wherein alveolar macrophages (AMs) play a central role. Dysfunctional mitochondria have been linked with distorted macrophages and hence lung disorders, but few treatments are currently available to correct these defects. Plant-derive nanovesicles have gained significant attention because of their therapeutic potential, but the targeting cells and the underlying mechanism remain elusive. We herein prepared the nanovesicles from Artemisia annua, a well-known medicinal plant with multiple attributes involving anti-inflammatory, anti-infection, and metabolism-regulating properties. By applying three mice models of acute lung injury caused by bacterial endotoxin, influenza A virus (IAV) and SARS-CoV-2 pseudovirus respectively, we showed that Artemisia-derived nanovesicles (ADNVs) substantially alleviated lung immunopathology and raised the survival rate of challenged mice. Macrophage depletion and adoptive transfer studies confirmed the requirement of AMs for ADNVs effects. We identified that gamma-aminobutyric acid (GABA) enclosed in the vesicles is a major molecular effector mediating the regulatory roles of ADNVs. Specifically, GABA acts on macrophages through GABA receptors, promoting mitochondrial gene programming and bioenergy generation, reducing oxidative stress and inflammatory signals, thereby enhancing the adaptability of AMs to inflammation resolution. Collectively, this study identifies a promising nanotherapeutics for alleviating lung pathology, and elucidates a mechanism whereby the canonical neurotransmitter modifies AMs and mitochondria to resume tissue homeostasis, which may have broader implications for treating critical pulmonary diseases such as COVID-19.


Asunto(s)
Lesión Pulmonar Aguda , Plantas Medicinales , Neumonía Viral , Neumonía , Ratones , Animales , Macrófagos Alveolares/metabolismo , Pulmón/metabolismo , Neumonía Viral/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Mitocondrias/patología , Ácido gamma-Aminobutírico/metabolismo , Neumonía/metabolismo
16.
J Nanobiotechnology ; 22(1): 464, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095755

RESUMEN

BACKGROUND: Doxorubicin (DOX) is a first-line chemotherapeutic drug for various malignancies that causes cardiotoxicity. Plant-derived exosome-like nanovesicles (P-ELNs) are growing as novel therapeutic agents. Here, we investigated the protective effects in DOX cardiotoxicity of ELNs from Momordica charantia L. (MC-ELNs), a medicinal plant with antioxidant activity. RESULTS: We isolated MC-ELNs using ultracentrifugation and characterized them with canonical mammalian extracellular vesicles features. In vivo studies proved that MC-ELNs ameliorated DOX cardiotoxicity with enhanced cardiac function and myocardial structure. In vitro assays revealed that MC-ELNs promoted cell survival, diminished reactive oxygen species, and protected mitochondrial integrity in DOX-treated H9c2 cells. We found that DOX treatment decreased the protein level of p62 through ubiquitin-dependent degradation pathway in H9c2 and NRVM cells. However, MC-ELNs suppressed DOX-induced p62 ubiquitination degradation, and the recovered p62 bound with Keap1 promoting Nrf2 nuclear translocation and the expressions of downstream gene HO-1. Furthermore, both the knockdown of Nrf2 and the inhibition of p62-Keap1 interaction abrogated the cardioprotective effect of MC-ELNs. CONCLUSIONS: Our findings demonstrated the therapeutic beneficials of MC-ELNs via increasing p62 protein stability, shedding light on preventive approaches for DOX cardiotoxicity.


Asunto(s)
Cardiotoxicidad , Doxorrubicina , Exosomas , Momordica charantia , Factor 2 Relacionado con NF-E2 , Animales , Cardiotoxicidad/prevención & control , Cardiotoxicidad/metabolismo , Momordica charantia/química , Exosomas/metabolismo , Ratas , Factor 2 Relacionado con NF-E2/metabolismo , Línea Celular , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Supervivencia Celular/efectos de los fármacos , Ratas Sprague-Dawley , Proteína Sequestosoma-1/metabolismo
17.
J Nanobiotechnology ; 22(1): 76, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38414031

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with different antigenic variants, has posed a significant threat to public health. It is urgent to develop inhalable vaccines, instead of injectable vaccines, to elicit mucosal immunity against respiratory viral infections. METHODS: We reported an inhalable hybrid nanovaccine (NVRBD-MLipo) to boost protective immunity against SARS-CoV-2 infection. Nanovesicles derived from genetically engineered 293T cells expressing RBD (NVRBD) were fused with pulmonary surfactant (PS)-biomimetic liposomes containing MPLA (MLipo) to yield NVRBD-MLipo, which possessed virus-biomimetic structure, inherited RBD expression and versatile properties. RESULTS: In contrast to subcutaneous vaccination, NVRBD-MLipo, via inhalable vaccination, could efficiently enter the alveolar macrophages (AMs) to elicit AMs activation through MPLA-activated TLR4/NF-κB signaling pathway. Moreover, NVRBD-MLipo induced T and B cells activation, and high level of RBD-specific IgG and secretory IgA (sIgA), thus elevating protective mucosal and systemic immune responses, while reducing side effects. NVRBD-MLipo also demonstrated broad-spectrum neutralization activity against SARS-CoV-2 (WT, Delta, Omicron) pseudovirus, and protected immunized mice against WT pseudovirus infection. CONCLUSIONS: This inhalable NVRBD-MLipo, as an effective and safe nanovaccine, holds huge potential to provoke robust mucosal immunity, and might be a promising vaccine candidate to combat respiratory infectious diseases, including COVID-19 and influenza.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Humanos , Ratones , Nanovacunas , COVID-19/prevención & control , Biomimética , Inmunidad Mucosa , Anticuerpos Antivirales , Anticuerpos Neutralizantes
18.
J Liposome Res ; 34(1): 135-177, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37144339

RESUMEN

Over the last few decades, cancer has been considered a clinical challenge, being among the leading causes of mortality all over the world. Although many treatment approaches have been developed for cancer, chemotherapy is still the most utilized in the clinical setting. However, the available chemotherapeutics-based treatments have several caveats including their lack of specificity, adverse effects as well as cancer relapse and metastasis which mainly explains the low survival rate of patients. Lipid nanoparticles (LNPs) have been utilized as promising nanocarrier systems for chemotherapeutics to overcome the challenges of the currently applied therapeutic strategies for cancer treatment. Loading chemotherapeutic agent(s) into LNPs improves drug delivery at different aspects including specific targeting of tumours, and enhancing the bioavailability of drugs at the tumour site through selective release of their payload, thus reducing their undesired side effects on healthy cells. This review article delineates an overview of the clinical challenges in many cancer treatments as well as depicts the role of LNPs in achieving optimal therapeutic outcomes. Moreover, the review contains a comprehensive description of the many LNPs categories used as nanocarriers in cancer treatment to date, as well as the potential of LNPs for future applications in other areas of medicine and research.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Liposomas , Neoplasias/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Lípidos , Portadores de Fármacos
19.
Nano Lett ; 23(17): 8013-8021, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37615624

RESUMEN

The rapid proliferative biological behavior of primary foci of anaplastic thyroid cancer (ATC) makes it a lethal tumor. According to the specific iodine uptake capacity of thyroid cells and enhanced endocytosis of ATC cells, we designed a kind of nanoclay drug-loading system and showed a promising treatment strategy for ATC. Introducing potassium iodide (KI) improves the homoaggregation of clay nanoparticles and then affects the distribution of nanoparticles in vivo, which makes KI@DOX-KaolinMeOH enriched almost exclusively in thyroid tissue. Simultaneously, the improvement of dispersibility of KI@DOX-KaolinMeOH changes the target uptake of ATC cells by improving the endocytosis and nanoparticle-induced autophagy, which regulate the production of autolysosomes and autophagy-enhanced chemotherapy, eventually contributing to a tumor inhibition rate of more than 90% in the primary foci of ATC. Therefore, this facile strategy to improve the homoaggregation of nanoclay by introducing KI has the potential to become an advanced drug delivery vehicle in ATC treatment.


Asunto(s)
Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides , Humanos , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Yoduro de Potasio/farmacología , Yoduro de Potasio/uso terapéutico , Caolín , Endocitosis , Sistemas de Liberación de Medicamentos , Neoplasias de la Tiroides/tratamiento farmacológico
20.
Nano Lett ; 23(10): 4101-4110, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37183806

RESUMEN

Osteoporosis is a global disease caused by abnormal overactivation of osteoclasts. The acidic environment in sealing zone of osteoclasts with H+ pumped from cytoplasm is critical to the maturation of osteoclasts. Therefore, reducing the intracellular H+ concentration can reduce the H+ secretion of osteoclasts from the source. In our study, we developed a novel nanovesicle which encapsulates Na2HPO4 with a liposome hybridizes with preosteoclast membrane (Na2HPO4@Lipo-pOCm). These nanovesicles release Na2HPO4 into the preosteoclast by targeting preosteoclasts and membrane fusion, reducing the intracellular H+ concentration, and achieve biological cascade regulation of osteoclasts through simple pH regulation. In vitro and in vivo experiments confirmed that these nanovesicles reduce mitochondrial membrane potential by decreasing intracellular H+ concentration, thereby reducing the ROS in osteoclasts as well as the expression of the upstream transcription factor FOXM1 of Acp5. In short, this nanovesicle can significantly inhibit the osteoclasts and ameliorate osteoporosis caused by OVX.


Asunto(s)
Osteoclastos , Osteoporosis , Humanos , Concentración de Iones de Hidrógeno , Homeostasis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda