Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Signal Transduct Target Ther ; 9(1): 89, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38616190

RESUMO

The inadequate tumor accumulation of anti-cancer agents is a major shortcoming of current therapeutic drugs and remains an even more significant concern in the clinical prospects for nanomedicines. Various strategies aiming at regulating the intratumoral permeability of therapeutic drugs have been explored in preclinical studies, with a primary focus on vascular regulation and stromal reduction. However, these methods may trigger or facilitate tumor metastasis as a tradeoff. Therefore, there is an urgent need for innovative strategies that boost intratumoral drug accumulation without compromising treatment outcomes. As another important factor affecting drug tumor accumulation besides vasculature and stroma, the impact of tumor-associated lymphatic vessels (LVs) has not been widely considered. In the current research, we verified that anlotinib, a tyrosine kinase inhibitor with anti-lymphangiogenesis activity, and SAR131675, a selective VEGFR-3 inhibitor, effectively decreased the density of tumor lymphatic vessels in mouse cancer models, further enhancing drug accumulation in tumor tissue. By combining anlotinib with therapeutic drugs, including doxorubicin (Dox), liposomal doxorubicin (Lip-Dox), and anti-PD-L1 antibody, we observed improved anti-tumor efficacy in comparison with monotherapy regimens. Meanwhile, this strategy significantly reduced tumor metastasis and elicited stronger anti-tumor immune responses. Our work describes a new, clinically transferrable approach to augmenting intratumoral drug accumulation, which shows great potential to address the current, unsatisfactory efficacies of therapeutic drugs without introducing metastatic risk.


Assuntos
Neoplasias , Animais , Camundongos , Neoplasias/tratamento farmacológico , Modelos Animais de Doenças , Nanomedicina
2.
J Biomed Mater Res A ; 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38487970

RESUMO

RNA-based therapeutics have gained traction for the prevention and treatment of a variety of diseases. However, their fragility and immunogenicity necessitate a drug carrier. Lipid nanoparticles (LNPs) have emerged as the predominant delivery vehicle for RNA therapeutics. An important component of LNPs is the ionizable lipid (IL), which is protonated in the acidic environment of the endosome, prompting cargo release into the cytosol. Currently, there is growing evidence that the structure of IL lipid tails significantly impacts the efficacy of LNP-mediated mRNA translation. Here, we optimized IL tail length for LNP-mediated delivery of three different mRNA cargos. Using C12-200, a gold standard IL, as a model, we designed a library of ILs with varying tail lengths and evaluated their potency in vivo. We demonstrated that small changes in lipophilicity can drastically increase or decrease mRNA translation. We identified that LNPs formulated with firefly luciferase mRNA (1929 base pairs) and C10-200, an IL with shorter tail lengths than C12-200, enhance liver transfection by over 10-fold. Furthermore, different IL tail lengths were found to be ideal for transfection of LNPs encapsulating mRNA cargos of varying sizes. LNPs formulated with erythropoietin (EPO), responsible for stimulating red blood cell production, mRNA (858 base pairs), and the C13-200 IL led to EPO translation at levels similar to the C12-200 LNP. The LNPs formulated with Cas9 mRNA (4521 base pairs) and the C9-200 IL induced over three times the quantity of indels compared with the C12-200 LNP. Our findings suggest that shorter IL tails may lead to higher transfection of LNPs encapsulating larger mRNAs, and that longer IL tails may be more efficacious for delivering smaller mRNA cargos. We envision that the results of this project can be utilized as future design criteria for the next generation of LNP delivery systems for RNA therapeutics.

3.
Proc Natl Acad Sci U S A ; 121(7): e2314747121, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38315853

RESUMO

Macrophages are integral components of the innate immune system, playing a dual role in host defense during infection and pathophysiological states. Macrophages contribute to immune responses and aid in combatting various infections, yet their production of abundant proinflammatory cytokines can lead to uncontrolled inflammation and worsened tissue damage. Therefore, reducing macrophage-derived proinflammatory cytokine release represents a promising approach for treating various acute and chronic inflammatory disorders. However, limited macrophage-specific delivery vehicles have hindered the development of macrophage-targeted therapies. In this study, we screened a pool of 112 lipid nanoparticles (LNPs) to identify an optimal LNP formulation for efficient siRNA delivery. Subsequently, by conjugating the macrophage-specific antibody F4/80 to the LNP surface, we constructed MacLNP, an enhanced LNP formulation designed for targeted macrophage delivery. In both in vitro and in vivo experiments, MacLNP demonstrated a significant enhancement in targeting macrophages. Specifically, delivery of siRNA targeting TAK1, a critical kinase upstream of multiple inflammatory pathways, effectively suppressed the phosphorylation/activation of NF-kB. LNP-mediated inhibition of NF-kB, a key upstream regulator in the classic inflammatory signaling pathway, in the murine macrophage cell line RAW264.7 significantly reduced the release of proinflammatory cytokines after stimulation with the viral RNA mimic Poly(I:C). Finally, intranasal administration of MacLNP-encapsulated TAK1 siRNA markedly ameliorated lung injury induced by influenza infection. In conclusion, our findings validate the potential of targeted macrophage interventions in attenuating inflammatory responses, reinforcing the potential of LNP-mediated macrophage targeting to treat pulmonary inflammatory disorders.


Assuntos
Lipossomos , Nanopartículas , Pneumonia Viral , Camundongos , Humanos , Animais , NF-kappa B/metabolismo , Lipídeos/farmacologia , Macrófagos/metabolismo , RNA Interferente Pequeno/metabolismo , Citocinas/metabolismo , Pneumonia Viral/metabolismo
4.
Nat Commun ; 15(1): 1884, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38424061

RESUMO

Lipid nanoparticles for delivering mRNA therapeutics hold immense promise for the treatment of a wide range of lung-associated diseases. However, the lack of effective methodologies capable of identifying the pulmonary delivery profile of chemically distinct lipid libraries poses a significant obstacle to the advancement of mRNA therapeutics. Here we report the implementation of a barcoded high-throughput screening system as a means to identify the lung-targeting efficacy of cationic, degradable lipid-like materials. We combinatorially synthesize 180 cationic, degradable lipids which are initially screened in vitro. We then use barcoding technology to quantify how the selected 96 distinct lipid nanoparticles deliver DNA barcodes in vivo. The top-performing nanoparticle formulation delivering Cas9-based genetic editors exhibits therapeutic potential for antiangiogenic cancer therapy within a lung tumor model in female mice. These data demonstrate that employing high-throughput barcoding technology as a screening tool for identifying nanoparticles with lung tropism holds potential for the development of next-generation extrahepatic delivery platforms.


Assuntos
DNA , Nanopartículas , Feminino , Animais , Camundongos , RNA Mensageiro/genética , Pulmão , Lipídeos
5.
Nat Commun ; 15(1): 1762, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409275

RESUMO

The ionizable lipidoid is a key component of lipid nanoparticles (LNPs). Degradable lipidoids containing extended alkyl branches have received tremendous attention, yet their optimization and investigation are underappreciated. Here, we devise an in situ construction method for the combinatorial synthesis of degradable branched (DB) lipidoids. We find that appending branch tails to inefficacious lipidoids via degradable linkers boosts mRNA delivery efficiency up to three orders of magnitude. Combinatorial screening and systematic investigation of two libraries of DB-lipidoids reveal important structural criteria that govern their in vivo potency. The lead DB-LNP demonstrates robust delivery of mRNA therapeutics and gene editors into the liver. In a diet-induced obese mouse model, we show that repeated administration of DB-LNP encapsulating mRNA encoding human fibroblast growth factor 21 alleviates obesity and fatty liver. Together, we offer a construction strategy for high-throughput and cost-efficient synthesis of DB-lipidoids. This study provides insights into branched lipidoids for efficient mRNA delivery.


Assuntos
Nanopartículas , Animais , Camundongos , Humanos , RNA Mensageiro/genética , Nanopartículas/química , RNA Interferente Pequeno
6.
J Control Release ; 362: 647-666, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37703928

RESUMO

Abnormal angiogenesis stands for one of the most striking manifestations of malignant tumor. The pathologically and structurally abnormal tumor vasculature facilitates a hostile tumor microenvironment, providing an ideal refuge exclusively for cancer cells. The emergence of vascular regulation drugs has introduced a distinctive class of therapeutics capable of influencing nutrition supply and drug delivery efficacy without the need to penetrate a series of physical barriers to reach tumor cells. Nanomedicines have been further developed for more precise regulation of tumor vasculature with the capacity of co-delivering multiple active pharmaceutical ingredients, which overall reduces the systemic toxicity and boosts the therapeutic efficacy of free drugs. Additionally, precise structure design enables the integration of specific functional motifs, such as surface-targeting ligands, droppable shells, degradable framework, or stimuli-responsive components into nanomedicines, which can improve tissue-specific accumulation, enhance tissue penetration, and realize the controlled and stimulus-triggered release of the loaded cargo. This review describes the morphological and functional characteristics of tumor blood vessels and summarizes the pivotal molecular targets commonly used in nanomedicine design, and then highlights the recent cutting-edge advancements utilizing nanotechnologies for precise regulation of tumor vasculature. Finally, the challenges and future directions of this field are discussed.

7.
Chem Commun (Camb) ; 59(72): 10745-10748, 2023 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-37581907

RESUMO

We propose a practical strategy to design a series of heavy-atom-free synergistic phototherapy agents (CSQs) with both photodynamic therapy (PDT) and photothermal therapy (PTT) under NIR wavelength excitation by simply replacing the indole salt of xanthene Changsha (CS) with quinoline salt.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Quinolinas , Humanos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia , Cloreto de Sódio , Neoplasias/tratamento farmacológico , Quinolinas/farmacologia
8.
Int J Mol Sci ; 24(11)2023 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-37298108

RESUMO

Although DNA damage repair plays a critical role in cancer chemotherapy, the function of lncRNAs in this process remains largely unclear. In this study, in silico screening identified H19 as an lncRNA that potentially plays a role in DNA damage response and sensitivity to PARP inhibitors. Increased expression of H19 is correlated with disease progression and with a poor prognosis in breast cancer. In breast cancer cells, forced expression of H19 promotes DNA damage repair and resistance to PARP inhibition, whereas H19 depletion diminishes DNA damage repair and increases sensitivity to PARP inhibitors. H19 exerted its functional roles via direct interaction with ILF2 in the cell nucleus. H19 and ILF2 increased BRCA1 stability via the ubiquitin-proteasome proteolytic pathway via the H19- and ILF2-regulated BRCA1 ubiquitin ligases HUWE1 and UBE2T. In summary, this study has identified a novel mechanism to promote BRCA1-deficiency in breast cancer cells. Therefore, targeting the H19/ILF2/BRCA1 axis might modulate therapeutic approaches in breast cancer.


Assuntos
Neoplasias da Mama , RNA Longo não Codificante , Humanos , Feminino , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/uso terapêutico , Ubiquitina/metabolismo , Dano ao DNA , Proteína do Fator Nuclear 45/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo
9.
Nat Nanotechnol ; 18(9): 1105-1114, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37365276

RESUMO

Lipid nanoparticle (LNP)-formulated messenger RNA (mRNA) vaccineare a promising platform to prevent infectious diseases as demonstrated by the recent success of SARS-CoV-2 mRNA vaccines. To avoid immune recognition and uncontrolled inflammation, nucleoside-modified mRNA is used. However, such modification largely abrogates the innate immune responses that are critical to orchestrating robust adaptive immunity. Here we develop an LNP component-an adjuvant lipidoid-that can enhance the adjuvanticity of mRNA-LNP vaccines. Our results show that partial substitution of ionizable lipidoid with adjuvant lipidoid not only enhanced mRNA delivery, but also endowed LNPs with Toll-like receptor 7/8-agonistic activity, which significantly increased the innate immunity of the SARS-CoV-2 mRNA-LNP vaccine with good tolerability in mice. Our optimized vaccine elicits potent neutralizing antibodies against multiple SARS-CoV-2 pseudovirus variants, strong Th1-biased cellular immunity, and robust B cell and long-lived plasma cell responses. Importantly, this adjuvant lipidoid substitution strategy works successfully in a clinically relevant mRNA-LNP vaccine, demonstrating its translational potential.


Assuntos
COVID-19 , Nanopartículas , Animais , Humanos , Camundongos , Vacinas contra COVID-19 , SARS-CoV-2/genética , COVID-19/prevenção & controle , Adjuvantes Imunológicos/farmacologia , RNA Mensageiro/genética , Vacinas de mRNA
10.
Environ Sci Pollut Res Int ; 30(18): 54149-54159, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36869175

RESUMO

In this study, the sludge-based biochar (BC) was prepared by dewatered sludge from a membrane bioreactor to treat the membrane concentrate. Then, the adsorbed and saturated BC was regenerated (RBC) by pyrolysis and deashing treatment to further treat the membrane concentrate. Afterward, the composition of membrane concentrate before and after BC or RBC treatment was detected, and the biochars' surface characteristics were characterized. The results showed that RBC outperformed BC in the abatement of chemical oxygen demand (CODCr), ammonia nitrogen (NH3-N), and total nitrogen (TN), with their removal rates of 60.07%, 51.55%, and 66.00%, respectively, an improvement of 9.49%, 9.00% and 16.50% of the removal rate compare to BC. The specific surface area of BC and RBC was about 109 times as much as the original dewatered sludge, and the pore size of BC and RBC belonged to mesopore which was a benefit for removing small and mediate size pollutants. The increase of the oxygen-containing functional group in RBC and the ash abatement contributed much to the improvement of RBC adsorption performance. In addition, cost analysis showed that BC+RBC had a cost of 0.76$/kg for COD removal, which was a lower cost than other common membrane concentrate treatment technologies.


Assuntos
Esgotos , Poluentes Químicos da Água , Poluentes Químicos da Água/análise , Adsorção , Carvão Vegetal/química , Nitrogênio/análise
11.
Int J Mol Sci ; 24(6)2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36982663

RESUMO

We investigated the expression and biological function of retinoic acid inducible gene I (RIG-I) in esophageal squamous cell carcinoma (ESCC). Materials and methods: An immunohistochemical analysis was performed on 86 pairs of tumor tissue and adjacent normal tissue samples of patients with ESCC. We generated RIG-I-overexpressing ESCC cell lines KYSE70 and KYSE450, and RIG-I- knockdown cell lines KYSE150 and KYSE510. Cell viability, migration and invasion, radioresistance, DNA damage, and cell cycle were evaluated using CCK-8, wound-healing and transwell assay, colony formation, immunofluorescence, and flow cytometry and Western blotting, respectively. RNA sequencing was performed to determine the differential gene expression between controls and RIG-I knockdown. Tumor growth and radioresistance were assessed in nude mice using xenograft models. RIG-I expression was higher in ESCC tissues compared with that in matched non-tumor tissues. RIG-I overexpressing cells had a higher proliferation rate than RIG-I knockdown cells. Moreover, the knockdown of RIG-I slowed migration and invasion rates, whereas the overexpression of RIG-I accelerated migration and invasion rates. RIG-I overexpression induced radioresistance and G2/M phase arrest and reduced DNA damage after exposure to ionizing radiations compared with controls; however, it silenced the RIG-I enhanced radiosensitivity and DNA damage, and reduced the G2/M phase arrest. RNA sequencing revealed that the downstream genes DUSP6 and RIG-I had the same biological function; silencing DUSP6 can reduce the radioresistance caused by the overexpression of RIG-I. RIG-I knockdown depleted tumor growth in vivo, and radiation exposure effectively delayed the growth of xenograft tumors compared with the control group. RIG-I enhances the progression and radioresistance of ESCC; therefore, it may be a new potential target for ESCC-targeted therapy.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Animais , Humanos , Camundongos , Carcinogênese/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Fosfatase 6 de Especificidade Dupla/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/radioterapia , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/genética , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Receptores do Ácido Retinoico/metabolismo
12.
Adv Mater ; : e2211609, 2023 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-36989141

RESUMO

Induced pluripotent stem cells (iPSCs) have wide applications in disease modeling, personalized medicine, and tissue engineering. The generation of iPSCs from somatic cells via transcriptional-factor- or chemical molecule-based approaches are time-consuming and inefficient. Here, a cell-reprogramming-inspired dynamically responsive hydrogel is fabricated via a synthetic-biology-based strategy. Human and mouse somatic cells (including senescent cells) are efficiently reprogrammed into iPSCs that exhibit key features of embryonic stem cells. The cell-reprogramming-responsive hydrogel possesses dynamic bioresponsiveness, and it faithfully senses metabolic remodeling and extracellular acidification during cell reprogramming, responding by changing its mechanical properties accordingly. Mechanistic study demonstrates that the autonomous change of the mechanical properties of the cell-reprogramming-responsive hydrogel elicits the formation of Yes-associated protein (YAP) biomolecular condensates with the appropriate timing during cell reprogramming, ensuring a faster and more efficient generation of iPSCs than conventional cell reprogramming approach. Taken together, this study reveals the robust induction of pluripotency by coordination of cell-reprogramming-inspired dynamically responsive hydrogel and phase-separated biomolecular condensates.

13.
Circ Res ; 132(3): 339-354, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36625267

RESUMO

BACKGROUND: During long-term antiplatelet agents (APAs) administration, patients with thrombotic diseases take a fairly high risk of life-threatening bleeding, especially when in need of urgent surgery. Rapid functional reversal of APAs remains an issue yet to be efficiently resolved by far due to the lack of any specific reversal agent in the clinic, which greatly restricts the use of APAs. METHODS: Flow cytometry analysis was first applied to assess the dose-dependent reversal activity of platelet-mimicking perfluorocarbon-based nanosponges (PLT-PFCs) toward ticagrelor. The tail bleeding time of mice treated with APAs followed by PLT-PFCs was recorded at different time points, along with corresponding pharmacokinetic analysis of ticagrelor and tirofiban. A hemorrhagic transformation model was established in experimental stroke mice with thrombolytic/antiplatelet therapy. Magnetic resonance imaging was subsequently applied to observe hemorrhage and thrombosis in vivo. Further evaluation of the spontaneous clot formation activity of PLT-PFCs was achieved by clot retraction assay in vitro. RESULTS: PLT-PFCs potently reversed the antiplatelet effect of APAs by competitively binding with APAs. PLT-PFCs showed high binding affinity comparable to fresh platelets in vitro with first-line APAs, ticagrelor and tirofiban, and efficiently reversed their function in both tail bleeding and postischemic-reperfusion models. Moreover, the deficiency of platelet intrinsic thrombotic activity diminished the risk of thrombogenesis. CONCLUSIONS: This study demonstrated the safety and effectiveness of platelet-mimicking nanosponges in ameliorating the bleeding risk of different APAs, which offers a promising strategy for the management of bleeding complications induced by antiplatelet therapy.


Assuntos
Inibidores da Agregação Plaquetária , Trombose , Animais , Camundongos , Inibidores da Agregação Plaquetária/efeitos adversos , Plaquetas , Ticagrelor/efeitos adversos , Tirofibana/efeitos adversos , Hemorragia/induzido quimicamente , Trombose/tratamento farmacológico , Trombose/prevenção & controle , Trombose/induzido quimicamente
14.
Adv Mater ; 35(3): e2207890, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36341495

RESUMO

Microbiota-based therapeutics offer innovative strategies to treat inflammatory bowel diseases (IBDs). However, the poor clinical outcome so far and the limited flexibility of the bacterial approach call for improvement. Inspired by the health benefits of probiotics in alleviating symptoms of bowel diseases, bioartificial probiotics are designed to restore the intestinal microenvironment in colitis by regulating redox balance, immune responses, and the gut microbiome. The bioartificial probiotic comprises two components: an E. coli Nissle 1917-derived membrane (EM) as the surface and the biodegradable diselenide-bridged mesoporous silica nanoparticles (SeM) as the core. When orally administered, the probiotic-inspired nanomedicine (SeM@EM) adheres strongly to the mucus layer and restored intestinal redox balance and immune regulation homeostasis in a murine model of acute colitis induced by dextran sodium sulfate. In addition, the respective properties of the EM and SeM synergistically alter the gut microbiome to a favorable state by increasing the bacterial diversity and shifting the microbiome profile to an anti-inflammatory phenotype. This work suggests a safe and effective nanomedicine that can restore intestinal homeostasis for IBDs therapy.


Assuntos
Colite , Microbioma Gastrointestinal , Animais , Camundongos , Nanomedicina , Escherichia coli/fisiologia , Colite/induzido quimicamente , Colite/tratamento farmacológico , Imunidade , Homeostase , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
15.
ACS Nano ; 16(11): 18253-18265, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36288552

RESUMO

Periodontitis is a chronic inflammatory disease caused by the interaction of oral microorganisms with the host immune response. Porphyromonas gingivalis (P.g.) acts as a key mediator in subverting the homeostasis of the local immune system. On the one hand, P.g. inhibits phagocytosis and the killing capacity of immune cells. On the other hand, P.g. increases selective cytokine release, which is beneficial to its further proliferation. Here, we prepared a penetrating macrophage-based nanoformulation (MZ@PNM)-encapsulating hydrogel (MZ@PNM@GCP) that responded to the periodontitis microenvironment. MZ@PNM targeted P.g. via the Toll-like receptor complex 2/1 (TLR2/1) on its macrophage-mimicking membrane, then directly killed P.g. through disruption of bacterial structural integrity by the cationic nanoparticles and intracellular release of an antibacterial drug, metronidazole (MZ). Meanwhile, MZ@PNM interrupted the specific binding of P.g. to immune cells and neutralized complement component 5a (C5a), preventing P.g. subversion of periodontal host immune response. Overall, MZ@PNM@GCP showed potent efficacy in periodontitis treatment, restoring local immune function and killing pathogenic bacteria, while exhibiting favorable biocompatibility, all of which have been demonstrated both in vivo and in vitro.


Assuntos
Periodontite , Humanos , Periodontite/tratamento farmacológico , Periodontite/metabolismo , Porphyromonas gingivalis/fisiologia , Macrófagos/metabolismo , Citocinas
17.
ACS Appl Mater Interfaces ; 14(31): 35366-35375, 2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-35914110

RESUMO

Gating systems have been extensively researched in energy harvesting, lab-on-chip applications, and so forth. However, the controlled drug delivery system with artificial hydrogel-based porous gating systems (HPGSs) is rarely reported. Herein, a biomimetic HPGS with a pH-responsive hydrogel as the valve and polydimethylsiloxane as the frame is fabricated by in situ femtosecond laser microdrilling and subsequent ultraviolet exposure. The proposed HPGS loaded with doxorubicin hydrochloride (DOX) is stable under physiological conditions, has a low drug leakage rate, and can achieve sustained drug release in a low pH environment. The experimental results show that the drug release is mainly controlled by non-Fickian diffusion, which renders the dynamic speed control of molecular transport possible. Moreover, the HPGS can also be prepared into an antitumor microcapsule. The results of in vitro cell experiments demonstrate that DOX@HPGS can release drugs and achieve terrific therapeutic efficacy in the elimination of HeLa cells in the acidic environments around tumor cells. This functional HPGS is envisioned to be an ideal pH-response carrier for sustained drug release treatment of digestive diseases such as inflammatory bowel disease and gastrointestinal cancer.


Assuntos
Doxorrubicina , Hidrogéis , Doxorrubicina/química , Doxorrubicina/farmacologia , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Células HeLa , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Concentração de Íons de Hidrogênio , Porosidade
18.
Polymers (Basel) ; 14(11)2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35683915

RESUMO

For the development of biodegradable superabsorbent polymers, the effect of the cross-linking length on the absorption characteristics of the Na salt of polyaspartic acid (PAspNa) was demonstrated using different concentrations of diamine cross-linking agents bearing carbon chains of different lengths, viz., ethylenediamine, 1,6-hexamethylenediamine, 1,8-diaminooctane, 1,10-diaminodecane, and 1,12-diaminododecane were used as cross-linking agents. The absorption of PAspNa was measured in deionised water and in a 0.9% aqueous NaCl solution. Under the conditions tested, when the alkyl chain of PAspNa was too short or too long, the absorbency was low and the cross-linking length was optimum. The success of the cross-linking reaction was confirmed by FT-IR spectroscopy. The degree of cross-linking was estimated and the ideal concentration for maximum water absorption was determined by elemental analysis. The sample obtained by cross-linking 1,8-diaminooctane at a concentration of 0.11 g/g polysuccinimide (PSI) showed the highest absorption. The thermal properties of each material were determined by dynamic scanning calorimetry. Therefore, the length of the cross-linking agent was found to strongly influence water absorption.

19.
ACS Cent Sci ; 7(11): 1938-1948, 2021 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-34841064

RESUMO

Polymeric nanoparticles have been extensively explored for biomedical applications, especially as framework materials for the construction of functional nanostructures. However, less attention has been paid to the inherent biological activities of those polymers. In this work, one of the commonly used polymers in gene and protein delivery, polyethylenimine-poly(lactic-co-glycolic acid)2 (PEI-PLGA), was discovered by accident to be able to mediate the nanoparticles to target the submandibular salivary glands of mice after intravenous injection. PEI-PLGA nanoparticles with an unmodified PEI surface selectively accumulated in submandibular salivary glands with ex vivo and in vitro study, suggesting that a ligand-receptor interaction between PEI and muscarinic acetylcholine receptor subtype 3 (M3 receptor) contributed to this affinity. Docking computation for the molecular binding mode between PEI segments and M3 receptor indicated the way they interacted was similar to that of the FDA-approved specific M3 receptor antagonist, tiotropium. The key amino acids mediated this specific interaction between PEI-PLGA nanoparticles and M3 receptor were identified via a simulated alanine mutation study. This work demonstrates the unique characteristic of PEI-PLGA nanoparticles, which may be useful for the development of muscarinic receptor targeted nanomedicines and should be taken into consideration when PEI-based nanoparticles are applied in gene delivery.

20.
Nano Lett ; 21(6): 2588-2595, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33650872

RESUMO

Compared with traditional chemotherapeutics, vascular disruption agents (VDAs) have the advantages of rapidly blocking the supply of nutrients and starving tumors to death. Although the VDAs are effective under certain scenarios, this treatment triggers angiogenesis in the later stage of therapy that frequently leads to tumor recurrence and treatment failure. Additionally, the nonspecific tumor targeting and considerable side effects also impede the clinical applications of VDAs. Here we develop a customized strategy that combines a VDA with an anti-angiogenic drug (AAD) using mesoporous silica nanoparticles (MSNs) coated with platelet membrane for the self-assembled tumor targeting accumulation. The tailor-made nanoparticles accumulate in tumor tissues through the targeted adhesion of platelet membrane surface to damaged vessel sites, resulting in significant vascular disruption and efficient anti-angiogenesis in animal models. This study demonstrates the promising potential of combining VDA and AAD in a single nanoplatform for tumor eradication.


Assuntos
Nanopartículas , Neoplasias , Inibidores da Angiogênese/uso terapêutico , Animais , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Dióxido de Silício/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...