Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 247
Filtrar
1.
J Inflamm Res ; 17: 5093-5112, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39099664

RESUMO

Background: Sepsis continues to exert a significant impact on morbidity and mortality in clinical settings, with immunosuppression, multi-organ failure, and disruptions in gut microbiota being key features. Although rheinic acid and tanshinone IIA show promise in mitigating macrophage apoptosis in sepsis treatment, their precise targeting of macrophages remains limited. Additionally, the evaluation of intestinal flora changes following treatment, which plays a significant role in subsequent cytokine storms, has been overlooked. Leveraging the innate inflammation chemotaxis of tumor cell-derived exosomes allows for their rapid recognition and uptake by activated macrophages, facilitating phenotypic changes and harnessing anti-inflammatory effects. Methods: We extracted exosomes from H1299 cells using a precipitation method. Then we developed a tumor cell-derived exosomal hybrid nanosystem loaded with rhubarbic acid and tanshinone IIA (R+T/Lipo/EXO) for sepsis treatment. In vitro studies, we verify the anti-inflammatory effect and the mechanism of inhibiting cell apoptosis of nano drug delivery system. The anti-inflammatory effects, safety, and modulation of intestinal microbiota by the nanoformulations were further validated in the in vivo study. Results: Nanoformulation demonstrated enhanced macrophage internalization, reduced TNF-α expression, inhibited apoptosis, modulated intestinal flora, and alleviated immunosuppression. Conclusion: R+T/Lipo/EXO presents a promising approach using exosomal hybrid nanosystems for treating sepsis.

2.
Int J Biol Macromol ; 278(Pt 1): 134661, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39128741

RESUMO

Hypoxia and high concentration of glutathione (GSH) in tumor seriously hinder the role of reactive oxygen species (ROS) and oxygen-dependence strategy in tumor treatment. In this work, a self-generating oxygen and self-consuming GSH hyaluronic acid (HA)-coated porphyrin nanoplatform (TAPPP@CaO2/Pt(IV)/HA) is established for enhancing photodynamic/ion/chemo targeting synergistic therapy of tumor. During the efforts of ROS production by nanosystems, a GSH consuming strategy is implemented for augmenting ROS-induced oxidative damage for synergetic cancer therapy. CaO2 in the nanosystems is decomposed into O2 and H2O2 in an acidic environment, which alleviates hypoxia and enhances the photodynamic therapy (PDT) effect. Calcium overload causes mitochondria dysfunction and induces apoptosis. Pt (IV) reacts with GSH to produce Pt (II) for chemotherapy and reduce the concentration of GSH, protecting ROS from scavenging for augmenting ROS-induced oxidative damage. In vitro and in vivo results demonstrated the self-generating oxygen and self-consuming GSH strategy can enhance ROS-dependent PDT coupled with ion/chemo synergistic therapy. The proposed strategy not only solves the long-term problem that hypoxia limits therapeutic effect of PDT, but also ameliorates the highly reducing environment of tumors. Thus the preparation of TAPPP@CaO2/Pt(IV)/HA provided a novel strategy for the effective combined therapy of cancers.

3.
Artigo em Inglês | MEDLINE | ID: mdl-39189605

RESUMO

In situ vaccines that can stimulate tumor immune response have emerged as a breakthrough in antitumor therapy. However, the immunosuppressed tumor microenvironment and insufficient infiltration of immune cells lead to ineffective antitumor immunity. Hence, a biomimetic carrier-free nanosystem (BCC) to induce synergistic phototherapy/chemotherapy-driven in situ vaccines was designed. A carrier-free nanosystem was developed using phototherapeutic reagents CyI and celastrol as raw materials. In vitro and in vivo studies have shown that under NIR light irradiation, BCC-mediated photo/chemotherapy not only accelerates the release of drugs to deeper parts of tumors, achieving timing and light-controlled drug delivery to result in cell apoptosis, but also effectively stimulates the antitumor response to induce in situ vaccine, which could invoke long-lasting antitumor immunity to inhibit tumor metastasis and eliminate distant tumor. This therapeutic strategy holds promise for priming robust innate and adaptive immune responses, arresting cancer progression, and inducing tumor dormancy.

4.
Artigo em Inglês | MEDLINE | ID: mdl-39136188

RESUMO

Recently, physical tools for remotely stimulating mechanical force-sensitive and temperature-sensitive proteins to regulate intracellular pathways have opened up novel and exciting avenues for basic research and clinical applications. Among the numerous modes of physical stimulation, magnetic stimulation is significantly attractive for biological applications due to the advantages of depth penetration and spatial-temporally controlled transduction. Herein, the physicochemical parameters (e.g., shape, size, composition) that influence the magnetic properties of magnetic nanosystems as well as the characteristics of transient receptor potential vanilloid-1 (TRPV1) and transient receptor potential vanilloid-4 (TRPV4) channels are systematically summarized, which offer opportunities for magnetic manipulation of cell fate in a precise and effective manner. In addition, representative regulatory applications involving magnetic nanosystem-based TRPV1 and TRPV4 channel activation are highlighted, both at the cellular level and in animal models. Furthermore, perspectives on the further development of this magnetic stimulation mode are commented on, with emphasis on scientific limitations and possible directions for exploitation. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.


Assuntos
Canais de Cátion TRPV , Canais de Cátion TRPV/metabolismo , Animais , Humanos , Camundongos
5.
Small ; : e2402502, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39007246

RESUMO

Inflammatory Bowel Disease (IBD) is a chronic inflammatory condition affecting the gastrointestinal tract (GIT). Glucagon-like peptide-2 (GLP-2) analogs possess high potential in the treatment of IBD by enhancing intestinal repair and attenuating inflammation. Due to the enzymatic degradation and poor intestinal absorption, GLP-2 analogs are administered parenterally, which leads to poor patient compliance. This work aims to develop IBD-targeted nanoparticles (NPs) for the oral delivery of the GLP-2 analog, Teduglutide (TED). Leveraging the overproduction of Reactive Oxygen Species (ROS) in the IBD environment, ROS-sensitive NPs are developed to target the intestinal epithelium, bypassing the mucus barrier. PEGylation of NPs facilitates mucus transposition, but subsequent PEG removal is crucial for cellular internalization. This de-PEGylation is possible by including a ROS-sensitive thioketal linker within the system. ROS-sensitive NPs are established, with the ability to fully de-PEGylate via ROS-mediated cleavage. Encapsulation of TED into NPs resulted in the absence of absorption in 3D in vitro models, potentially promoting a localized action, and avoiding adverse effects due to systemic absorption. Upon oral administration to colitis-induced mice, ROS-sensitive NPs are located in the colon, displaying healing capacity and reducing inflammation. Cleavable PEGylated NPs demonstrate effective potential in managing IBD symptoms and modulating the disease's progression.

6.
Heliyon ; 10(13): e33837, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39050425

RESUMO

Because of the tumor's recurrence and significant metastasis, the standard single-therapy paradigm has failed to meet clinical requirements. Recently, researchers have focused their emphasis on phototherapy and immunogenic cell death (ICD) techniques. In response to the current problems of immunotherapy, a multifunctional drug delivery nanosystem (PDA-IMQ@CaCO3-blinatumomab, PICB) was constructed by using high physiological compatibility of polydopamine (PDA) and calcium carbonate (CaCO3). Toll-like receptor 7 (TLR7) agonist imiquimod (IMQ) and bispecific antibody (BsAb) blinatumomab were loaded onto PDA-CaCO3 nanoparticles (NPs). The findings revealed that the system exhibited the advantages of good dispersion, high stability, excellent physiological compatibility, low toxicity, and high drug loading rate. Compared to the control group, it resulted in a 2.4-fold decrease in FOXP3+ regulatory T-cells within the tumor and a 5.0-fold increase in CD4+ effector T-cells, and promoted the production of damage-related molecular patterns to reinvigorate the ICD effect. PICB had a strong inhibitory effect on tumor growth in 4T1 tumor-bearing mice, and has no toxicity to other organs. Therefore, the multifunctional drug delivery nanosystem constructed in this study could effectively exert the properties of various components in vivo, fully demonstrate the synergistic effect between immunotherapy and photothermal therapy, thus significantly improving the tumor therapeutic efficacy, and has a promising clinical application.

7.
Int J Nanomedicine ; 19: 5605-5618, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38882547

RESUMO

Patients diagnosed with glioma typically face a limited life expectancy (around 15 months on average), a bleak prognosis, and a high likelihood of recurrence. As such, glioma is recognized as a significant form of malignancy. Presently, the treatment options for glioma include traditional approaches such as surgery, chemotherapy, and radiotherapy. Regrettably, the efficacy of these treatments has been less than optimal. Nevertheless, a promising development in glioma treatment lies in the use of hydrogel nano-systems as sophisticated delivery systems. These nano-systems have demonstrated exceptional therapeutic effects in the treatment of glioma by various responsive ways, including temperature-response, pH-response, liposome-response, ROS-response, light-response, and enzyme-response. This study seeks to provide a comprehensive summary of both the therapeutic application of hydrogel nano-systems in managing glioma and the underlying immune action mechanisms.


Assuntos
Neoplasias Encefálicas , Glioma , Hidrogéis , Glioma/terapia , Humanos , Hidrogéis/química , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/tratamento farmacológico , Animais , Lipossomos/química , Sistemas de Liberação de Medicamentos/métodos , Nanomedicina , Nanopartículas/química
8.
Pharmacol Rep ; 76(4): 823-837, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38888724

RESUMO

BACKGROUND: Traditional small-molecule chemotherapeutics usually do not distinguish tumors from healthy tissues. However, nanotechnology creates nanocarriers that selectively deliver drugs to their site of action. This work is the next step in the development of the quantum dot-ß-cyclodextrin-folic acid (QD-ß-CD-FA) platform for targeted and selected delivery of C-2028 unsymmetrical bisacridine in cancer therapy. METHODS: Herein, we report an initial biological evaluation (using flow cytometry and light microscopy) as well as cell migration analysis of QD-ß-CD(C-2028)-FA nanoconjugate and its components in the selected human lung and prostate cancer cells, as well as against their respective normal cells. RESULTS: C-2028 compound induced apoptosis, which was much stronger in cancer cells compared to normal cells. Conjugation of C-2028 with QDgreen increased cellular senescence, while the introduction of FA to the conjugate significantly decreased this process. C-2028 nanoencapsulation also reduced cell migration. Importantly, QDgreen and QDgreen-ß-CD-FA themselves did not induce any toxic responses in studied cells. CONCLUSIONS: In conclusion, the results demonstrate the high potential of a novel folic acid-targeted receptor quantum dot-ß-cyclodextrin carrier (QDgreen-ß-CD-FA) for drug delivery in cancer treatment. Nanoplatforms increased the amount of delivered compounds and demonstrated high suitability.


Assuntos
Apoptose , Portadores de Fármacos , Ácido Fólico , Neoplasias Pulmonares , Neoplasias da Próstata , Pontos Quânticos , beta-Ciclodextrinas , Humanos , Masculino , beta-Ciclodextrinas/química , Ácido Fólico/química , Ácido Fólico/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Pontos Quânticos/química , Apoptose/efeitos dos fármacos , Portadores de Fármacos/química , Movimento Celular/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Acridinas/farmacologia , Acridinas/administração & dosagem , Acridinas/química , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos
9.
Biomaterials ; 311: 122678, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38917705

RESUMO

Drug transmission through the blood-brain barrier (BBB) is considered an arduous challenge for brain injury treatment following the return of spontaneous circulation after cardiac arrest (CA-ROSC). Inspired by the propensity of melanoma metastasis to the brain, B16F10 cell membranes are camouflaged on 2-methoxyestradiol (2ME2)-loaded reactive oxygen species (ROS)-triggered "Padlock" nanoparticles that are constructed by phenylboronic acid pinacol esters conjugated D-a-tocopheryl polyethylene glycol succinate (TPGS-PBAP). The biomimetic nanoparticles (BM@TP/2ME2) can be internalized, mainly mediated by the mutual recognition and interaction between CD44v6 expressed on B16F10 cell membranes and hyaluronic acid on cerebral vascular endothelial cells, and they responsively release 2ME2 by the oxidative stress microenvironment. Notably, BM@TP/2ME2 can scavenge excessive ROS to reestablish redox balance, reverse neuroinflammation, and restore autophagic flux in damaged neurons, eventually exerting a remarkable neuroprotective effect after CA-ROSC in vitro and in vivo. This biomimetic drug delivery system is a novel and promising strategy for the treatment of cerebral ischemia-reperfusion injury after CA-ROSC.


Assuntos
2-Metoxiestradiol , Parada Cardíaca , Nanopartículas , Espécies Reativas de Oxigênio , Animais , Espécies Reativas de Oxigênio/metabolismo , Nanopartículas/química , Camundongos , 2-Metoxiestradiol/farmacologia , 2-Metoxiestradiol/química , Parada Cardíaca/tratamento farmacológico , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Masculino , Camundongos Endogâmicos C57BL , Sistemas de Liberação de Medicamentos , Linhagem Celular Tumoral , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Estresse Oxidativo/efeitos dos fármacos
10.
Nanomaterials (Basel) ; 14(12)2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38921898

RESUMO

A new curcuminoid molecule (3) has been designed and synthesized, containing a central -(CH2)2-COOH chain at the α carbon of the keto-enol moiety in the structure. The carboxylic acid group is added to react with exposed amino groups on silica oxide nanoparticles (nSiO2), forming an amide bond to attach the curcuminoid moiety to the nSiO2 covalently. The Kaiser test quantifies the functionalization degree, yielding 222 µmol of curcuminoid per gram of nanoparticles. The synthesized hybrid nanosystem, nSiO2-NHCO-CCM, displays significant emission properties, with a maximum emission at 538 nm in dichloromethane, similar to curcuminoid 1 (without the central chain), which emits at 565 nm in the same solvent. Solvent-induced spectral effects on the absorption and emission bands of the new hybrid nanosystem are confirmed, similar to those observed for the free curcuminoid (1). The new nanosystem is evaluated in the presence of kerosene in water, showing an emission band at 525 nm as a detection response. The ability of nSiO2-NHCO-CCM to change its fluorescence when interacting with kerosene in water is notable, as it overcomes the limitation caused by the insolubility of free curcuminoid 1 in water, allowing for the exploitation of its properties when connected to the water-stable nanosystem for future detection studies.

11.
Exploration (Beijing) ; 4(3): 20230048, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38939864

RESUMO

The fibrillation of amyloid-ß (Aß) is the critical causal factor in Alzheimer's disease (AD), the dissolution and clearance of which are promising for AD therapy. Although many Aß inhibitors are developed, their low Aß-binding affinity results in unsatisfactory effect. To solve this challenge, the Aß sequence-matching strategy is proposed to tail-design dissociable nanosystem (B6-PNi NPs). Herein, B6-PNi NPs aim to improve Aß-binding affinity for effective dissolution of amyloid fibrils, as well as to interfere with the in vivo fate of amyloid for Aß clearance. Results show that B6-PNi NPs decompose into small nanostructures and expose Aß-binding sites in response to AD microenvironment, and then capture Aß via multiple interactions, including covalent linkage formed by nucleophilic substitution reaction. Such high Aß-binding affinity disassembles Aß fibrils into Aß monomers, and induces the reassembly of Aß&nanostructure composite, thereby promoting microglial Aß phogocytosis/clearance via Aß receptor-mediated endocytosis. After B6-PNi NPs treatment, the Aß burden, neuroinflammation and cognitive impairments are relieved in AD transgenic mice. This work provides the Aß sequence-matching strategy for Aß inhibitor design in AD treatment, showing meaningful insight in biomedicine.

12.
Exp Biol Med (Maywood) ; 249: 10055, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38774281

RESUMO

Currently, various functionalized nanocarrier systems are extensively studied for targeted delivery of drugs, peptides, and nucleic acids. Joining the approaches of genetic and chemical engineering may produce novel carriers for precise targeting different cellular proteins, which is important for both therapy and diagnosis of various pathologies. Here we present the novel nanocontainers based on vectorized genetically encoded Myxococcus xanthus (Mx) encapsulin, confining a fluorescent photoactivatable mCherry (PAmCherry) protein. The shells of such encapsulins were modified using chemical conjugation of human transferrin (Tf) prelabeled with a fluorescein-6 (FAM) maleimide acting as a vector. We demonstrate that the vectorized encapsulin specifically binds to transferrin receptors (TfRs) on the membranes of mesenchymal stromal/stem cells (MSCs) followed by internalization into cells. Two spectrally separated fluorescent signals from Tf-FAM and PAmCherry are clearly distinguishable and co-localized. It is shown that Tf-tagged Mx encapsulins are internalized by MSCs much more efficiently than by fibroblasts. It has been also found that unlabeled Tf effectively competes with the conjugated Mx-Tf-FAM formulations. That indicates the conjugate internalization into cells by Tf-TfR endocytosis pathway. The developed nanoplatform can be used as an alternative to conventional nanocarriers for targeted delivery of, e.g., genetic material to MSCs.


Assuntos
Células-Tronco Mesenquimais , Myxococcus xanthus , Transferrina , Células-Tronco Mesenquimais/metabolismo , Transferrina/metabolismo , Humanos , Myxococcus xanthus/metabolismo , Endocitose , Receptores da Transferrina/metabolismo , Proteínas Luminescentes/metabolismo , Proteínas Luminescentes/genética
13.
ACS Biomater Sci Eng ; 10(6): 3994-4008, 2024 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-38736179

RESUMO

Disruption of the symbiosis of extra/intratumoral metabolism is a good strategy for treating tumors that shuttle resources from the tumor microenvironment. Here, we report a precision treatment strategy for enhancing pyruvic acid and intratumoral acidosis to destroy tumoral metabolic symbiosis to eliminate tumors; this approach is based on PEGylated gold and lactate oxidase-modified aminated dendritic mesoporous silica with lonidamine and ferrous sulfide loading (PEG-Au@DMSNs/FeS/LND@LOX). In the tumor microenvironment, LOX oxidizes lactic acid to produce pyruvate, which represses tumor cell proliferation by inhibiting histone gene expression and induces ferroptosis by partial histone monoubiquitination. In acidic tumor conditions, the nanoparticles release H2S gas and Fe2+ ions, which can inhibit catalase activity to promote the Fenton reaction of Fe2+, resulting in massive ·OH production and ferroptosis via Fe3+. More interestingly, the combination of H2S and LND (a monocarboxylic acid transporter inhibitor) can cause intracellular acidosis by lactate, and protons overaccumulate in cells. Multiple intracellular acidosis is caused by lactate-pyruvate axis disorders. Moreover, H2S provides motive power to intensify the shuttling of nanoparticles in the tumor region. The findings confirm that this nanomedicine system can enable precise antitumor effects by disrupting extra/intratumoral metabolic symbiosis and inducing ferroptosis and represents a promising active drug delivery system candidate for tumor treatment.


Assuntos
Ferroptose , Ácido Láctico , Ácido Pirúvico , Microambiente Tumoral , Ferroptose/efeitos dos fármacos , Humanos , Ácido Láctico/metabolismo , Animais , Ácido Pirúvico/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/terapia , Linhagem Celular Tumoral , Camundongos , Ouro/química , Dióxido de Silício/química , Feminino , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Camundongos Endogâmicos BALB C , Proliferação de Células/efeitos dos fármacos , Oxigenases de Função Mista , Indazóis
14.
Int J Biol Macromol ; 270(Pt 2): 132268, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38734336

RESUMO

Paclitaxel (PTX) is a potent anticancer drug. However, PTX exhibits extremely poor solubility in aqueous solution along with severe side effects. Therefore, in this study, an inclusion complex was prepared between PTX and hydroxypropyl-ß-cyclodextrin (HPßCD) by solvent evaporation to enhance the drug's solubility. The HPßCD-PTX inclusion complex was then encapsulated in poly-3-hydroxybutyrate (PHB) to fabricate drug-loaded nanoparticles (HPßCD-PTX/PHB NPs) by nanoprecipitation. The HPßCD-PTX/PHB NPs depicted a higher release of PTX at pH 5.5 thus demonstrating a pH-dependent release profile. The cytotoxic properties of HPßCD-PTX/PHB NPs were tested against MCF-7, MDA-MB-231 and SW-620 cell lines. The cytotoxic potential of HPßCD-PTX/PHB NPs was 2.59-fold improved in MCF-7 cells in comparison to free PTX. Additionally, the HPßCD-PTX/PHB NPs improved the antimitotic (1.68-fold) and apoptotic (8.45-fold) effects of PTX in MCF-7 cells in comparison to PTX alone. In summary, these pH-responsive nanoparticles could be prospective carriers for enhancing the cytotoxic properties of PTX for the treatment of breast cancer.


Assuntos
2-Hidroxipropil-beta-Ciclodextrina , Apoptose , Portadores de Fármacos , Nanopartículas , Paclitaxel , Poliésteres , Proibitinas , Humanos , Nanopartículas/química , Paclitaxel/farmacologia , Paclitaxel/química , Concentração de Íons de Hidrogênio , Apoptose/efeitos dos fármacos , 2-Hidroxipropil-beta-Ciclodextrina/química , Portadores de Fármacos/química , Poliésteres/química , Células MCF-7 , Hidroxibutiratos/química , Hidroxibutiratos/farmacologia , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Solubilidade , Sobrevivência Celular/efeitos dos fármacos , Poli-Hidroxibutiratos
15.
Adv Healthc Mater ; : e2400538, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38759954

RESUMO

Osteosarcoma (OS) is a "cold" tumor enriched in noninflammatory M2 phenotype tumor-associated macrophages (TAMs), which limits the efficacy of immunotherapy. The acidic tumor microenvironment (TME), generated by factors such as excess hydrogen (H+) ions and high lactate levels, activates immunosuppressive cells, further promoting a suppressive tumor immune microenvironment (TIME). Therefore, a multitarget synergistic combination strategy that neutralizes the acidic TME and reprograms TAMs can be beneficial for OS therapy. Here, a calcium carbonate (CaCO3)/polydopamine (PDA)-based nanosystem (A-NPs@(SHK+Ce6)) is developed. CaCO3 nanoparticles are used to neutralize H+ ions and alleviate the suppressive TIME, and the loaded SHK not only synergizes with photodynamic therapy (PDT) but also inhibits lactate production, further reversing the acidic TME and repolarizing TAMs to consequently lead to enhanced PDT-induced tumor suppression and comprehensive beneficial effects on antitumor immune responses. Importantly, A-NPs@(SHK+Ce6), in combination with programmed cell death protein 1 (PD-1) checkpoint blockade, shows a remarkable ability to eliminate distant tumors and promote long-term immune memory function to protect against rechallenged tumors. This work presents a novel multiple-component combination strategy that coregulates the acidic TME and TAM polarization to reprogram the TIME.

16.
Biomed Pharmacother ; 175: 116699, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38705129

RESUMO

Osteoporosis (OP) constitutes a significant health concern that profoundly affects individuals' quality of life. Bisphosphonates, conventional pharmaceuticals widely employed in OP treatment, encounter limitations related to inadequate drug targeting and a short effective duration, thereby compromising their clinical efficacy. The burgeoning field of nanotechnology has witnessed the development and application of diverse functional nanosystems designed for OP treatment. Owing to the bone tissue affinity of bisphosphonates, these nanosystems are modified to address shortcomings associated with traditional drug delivery. In this review, we explore the potential of bisphosphonate-modified nanosystems as a promising strategy for addressing osteoporotic conditions. With functional modification, these nanosystems exhibit a targeted and reversible effect on osteoporotic remodeling, presenting a promising solution to enhance precision in drug delivery. The synthesis methods, physicochemical properties, and in vitro/in vivo performance of bisphosphonate-modified nanosystems are comprehensively examined in this review. Through a thorough analysis of recent advances and accomplishments in this field, we aim to provide insights into the potential applications and future directions of bisphosphonate-modified nanosystems for targeted and reversible osteoporotic remodeling.


Assuntos
Conservadores da Densidade Óssea , Difosfonatos , Osteoporose , Humanos , Osteoporose/tratamento farmacológico , Difosfonatos/química , Difosfonatos/administração & dosagem , Animais , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/química , Conservadores da Densidade Óssea/farmacologia , Sistemas de Liberação de Medicamentos , Nanopartículas/química
17.
Small ; : e2400413, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38721946

RESUMO

Liver fibrosis is a coordinated response to tissue injury that is mediated by immune cell interactions. A mitochondria-regulated information-processing (MIP) nanosystem that promotes immune cell communication and interactions to inhibit liver fibrosis is designed. The MIP nanosystem mimics the alkaline amino acid domain of mitochondrial precursor proteins, providing precise targeting of the mitochondria. The MIP nanosystem is driven by light to modulate the mitochondria of hepatic stellate cells, resulting in the release of mitochondrial DNA into the fibrotic microenvironment, as detected by macrophages. By activating the STING signaling pathway, the developed nanosystem-induced macrophage phenotype switches to a reparative subtype (Ly6Clow) and downstream immunostimulatory transcriptional activity, fully restoring the fibrotic liver to its normal tissue state. The MIP nanosystem serves as an advanced information transfer system, allowing precise regulation of trained immunity, and offers a promising approach for effective liver fibrosis immunotherapy with the potential for clinical translation.

18.
Front Med (Lausanne) ; 11: 1385123, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38784236

RESUMO

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized primarily by synovitis, leading to the destruction of articular cartilage and bone and ultimately resulting in joint deformity, loss of function, and a significant impact on patients' quality of life. Currently, a combination of anti-rheumatic drugs, hormonal drugs, and biologics is used to mitigate disease progression. However, conventional drug therapy has limited bioavailability, and long-term use often leads to drug resistance and toxic side effects. Therefore, exploring new therapeutic approaches for RA is of great clinical importance. Nanodrug delivery systems offer promising solutions to overcome the limitations of conventional drugs. Among them, liposomes, the first nanodrug delivery system to be approved for clinical application and still widely studied, demonstrate the ability to enhance therapeutic efficacy with fewer adverse effects through passive or active targeting mechanisms. In this review, we provide a review of the research progress on the targeting mechanisms of various natural biomimetic nano-delivery systems in RA therapy. Additionally, we predict the development trends and application prospects of these systems, offering new directions for precision treatment of RA.

19.
Molecules ; 29(10)2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38792086

RESUMO

Photodynamic therapy (PDT) is a non-invasive anticancer treatment that uses special photosensitizer molecules (PS) to generate singlet oxygen and other reactive oxygen species (ROS) in a tissue under excitation with red or infrared light. Though the method has been known for decades, it has become more popular recently with the development of new efficient organic dyes and LED light sources. Here we introduce a ternary nanocomposite: water-soluble star-like polymer/gold nanoparticles (AuNP)/temoporfin PS, which can be considered as a third-generation PDT system. AuNPs were synthesized in situ inside the polymer molecules, and the latter were then loaded with PS molecules in an aqueous solution. The applied method of synthesis allows precise control of the size and architecture of polymer nanoparticles as well as the concentration of the components. Dynamic light scattering confirmed the formation of isolated particles (120 nm diameter) with AuNPs and PS molecules incorporated inside the polymer shell. Absorption and photoluminescence spectroscopies revealed optimal concentrations of the components that can simultaneously reduce the side effects of dark toxicity and enhance singlet oxygen generation to increase cancer cell mortality. Here, we report on the optical properties of the system and detailed mechanisms of the observed enhancement of the phototherapeutic effect. Combinations of organic dyes with gold nanoparticles allow significant enhancement of the effect of ROS generation due to surface plasmonic resonance in the latter, while the application of a biocompatible star-like polymer vehicle with a dextran core and anionic polyacrylamide arms allows better local integration of the components and targeted delivery of the PS molecules to cancer cells. In this study, we demonstrate, as proof of concept, a successful application of the developed PDT system for in vitro treatment of triple-negative breast cancer cells under irradiation with a low-power LED lamp (660 nm). We consider the developed nanocomposite to be a promising PDT system for application to other types of cancer.


Assuntos
Resinas Acrílicas , Ouro , Nanopartículas Metálicas , Fotoquimioterapia , Fármacos Fotossensibilizantes , Ouro/química , Fotoquimioterapia/métodos , Nanopartículas Metálicas/química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Humanos , Resinas Acrílicas/química , Linhagem Celular Tumoral , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Porfirinas/química , Porfirinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Polímeros/química , Antineoplásicos/farmacologia , Antineoplásicos/química
20.
Nano Lett ; 24(20): 6174-6182, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38739468

RESUMO

Accumulated reactive oxygen species (ROS) and their resultant vascular dysfunction in androgenic alopecia (AGA) hinder hair follicle survival and cause permanent hair loss. However, safe and effective strategies to rescue hair follicle viability to enhance AGA therapeutic efficiency remain challenging. Herein, we fabricated a quercetin-encapsulated (Que) and polydopamine-integrated (PDA@QLipo) nanosystem that can reshape the perifollicular microenvironment to initial hair follicle regeneration for AGA treatment. Both the ROS scavenging and angiogenesis promotion abilities of PDA@QLipo were demonstrated. In vivo assays revealed that PDA@QLipo administrated with roller-microneedles successfully rejuvenated the "poor" perifollicular microenvironment, thereby promoting cell proliferation, accelerating hair follicle renewal, and facilitating hair follicle recovery. Moreover, PDA@QLipo achieved a higher hair regeneration coverage of 92.5% in the AGA mouse model than minoxidil (87.8%), even when dosed less frequently. The nanosystem creates a regenerative microenvironment by scavenging ROS and augmenting neovascularity for hair regrowth, presenting a promising approach for AGA clinical treatment.


Assuntos
Alopecia , Folículo Piloso , Indóis , Polímeros , Quercetina , Espécies Reativas de Oxigênio , Alopecia/tratamento farmacológico , Alopecia/patologia , Quercetina/farmacologia , Quercetina/administração & dosagem , Quercetina/química , Animais , Indóis/química , Indóis/farmacologia , Folículo Piloso/efeitos dos fármacos , Folículo Piloso/crescimento & desenvolvimento , Polímeros/química , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Regeneração/efeitos dos fármacos , Humanos , Cabelo/efeitos dos fármacos , Cabelo/crescimento & desenvolvimento , Proliferação de Células/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Modelos Animais de Doenças , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA