Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 63(10): e202317943, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38078895

RESUMO

Metal-based drugs currently dominate the field of chemotherapeutic agents; however, achieving the controlled activation of metal prodrugs remains a substantial challenge. Here, we propose a universal strategy for the radiation-triggered activation of metal prodrugs via nanosurface energy transfer (NSET). The core-shell nanoplatform (Ru-GNC) is composed of gold nanoclusters (GNC) and ruthenium (Ru)-containing organic-inorganic hybrid coatings. Upon X-ray irradiation, chemotherapeutic Ru (II) complexes were released in a controlled manner through a unique NSET process involving the transfer of photoelectron energy from the radiation-excited Ru-GNCs to the Ru-containing hybrid layer. In contrast to the traditional radiation-triggered activation of prodrugs, such an NSET-based system ensures that the reactive species in the tumor microenvironment are present in sufficient quantity and are not easily quenched. Additionally, ultrasmall Ru-GNCs preferably target mitochondria and profoundly disrupt the respiratory chain upon irradiation, leading to radiosensitization by generating abundant reactive oxygen species. Consequently, Ru-GNC-directed radiochemotherapy induces immunogenic cell death, resulting in significant therapeutic outcomes when combined with the programmed cell death-ligand 1 (PD-L1) checkpoint blockade. This NSET strategy represents a breakthrough in designing radiation-triggered nanoplatforms for metal-prodrug-mediated cancer treatment in an efficient and controllable manner.


Assuntos
Pró-Fármacos , Pró-Fármacos/farmacologia , Transferência de Energia , Espécies Reativas de Oxigênio , Imunoterapia , Linhagem Celular Tumoral
2.
Small ; 19(14): e2207029, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36703529

RESUMO

The establishment of effective antitumor immune responses of vaccines is mainly limited by insufficient priming tumor infiltration of T cells and immunosuppressive tumor microenvironment (TME). Targeting ß-adrenergic receptor (ß-AR) signaling exerts promising benefits on reversing the suppressive effects directly on T cells, but it appears to have considerably limited antitumor performance when combined with vaccine-based immunotherapies. Herein, a tumor membrane-coated nanoplatform for codelivery of adjuvant CpG and propranolol (Pro), a ß-AR inhibitor is designed. The biomimetic nanovaccine displayed an improved accumulation in lymph nodes and sufficient drug release, thereby inducing dendritic cell maturation and antigen presentation. Meanwhile, the integration of vaccination and blockade of ß-AR signaling not only promoted the priming of the naive CD8+ T cells and effector T cell egress from lymph nodes, but also alleviated the immunosuppressive TME by decreasing the frequency of immunosuppressive cells and increasing the tumor infiltration of B cells and NK cells. Consequently, the biomimetic nanovaccines outperformed greater prophylactic and therapeutic efficacy than nanovaccines without Pro encapsulation in B16-F10 melanoma mice. Taken together, the work explored a biomimetic nanovaccine for priming tumor infiltration of T cells and immunosuppressive TME regulation, offering tremendous potential for a combined ß-AR signaling-targeting strategy in cancer immunotherapy.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Animais , Camundongos , Receptores Adrenérgicos beta , Biomimética , Imunoterapia , Transdução de Sinais , Neoplasias/tratamento farmacológico , Propranolol/farmacologia , Camundongos Endogâmicos C57BL , Células Dendríticas , Microambiente Tumoral
3.
J Nanobiotechnology ; 21(1): 235, 2023 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-37481565

RESUMO

Systemic administration of platinum-based drugs has obvious limitations in the treatment of advanced bladder cancer (BC) owing to lower tumor accumulation and uncontrolled release of chemotherapeutics. There is an urgent need for advanced strategies to overcome the current limitations of platinum-based chemotherapy, to achieve maximal therapeutic outcomes with reduced side effects. In this study, self-polymerized platinum (II)-polydopamine nanocomplexes (PtPDs) were tailored for efficient chemo-photoimmunotherapy of BC. PtPDs with high Pt loading content (11.3%) were degradable under the combination of a reductive tumor microenvironment and near-infrared (NIR) light irradiation, thus controlling the release of Pt ions to achieve efficient chemotherapy. In addition, polydopamine promoted stronger photothermal effects to supplement platinum-based chemotherapy. Consequently, PtPDs provided effective chemo-photothermal therapy of MB49 BC in vitro and in vivo, strengthening the immunogenic cell death (ICD) effect and robust anti-tumoral immunity response. When combined with a PD-1 checkpoint blockade, PtPD-based photochemotherapy evoked systemic immune responses that completely suppressed primary and distant tumor growth without inducing systemic toxicities. Our work provides a highly versatile approach through metal-dopamine self-polymerization for the precise delivery of metal-based chemotherapeutic drugs, and may serve as a promising nanomedicine for efficient and safe platinum-based chemotherapy for BC.


Assuntos
Nanomedicina , Neoplasias da Bexiga Urinária , Humanos , Polimerização , Indóis , Microambiente Tumoral
4.
Chem Rec ; 22(4): e202100287, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35020240

RESUMO

The integration of silver nanoparticles (Ag NPs) with mesoporous silica nanoparticles (MSNs) protects the former from aggregation and promotes the controlled release of silver ions, resulting in therapeutic significance on cancer and infection. The unique size, shape, pore structure and silver distribution of silver mesoporous silica nanoparticles (Ag-MSNs) embellish them with the potential to perform combined imaging and therapeutic actions via modulating optical and drug release properties. Here, we comprehensively review the recent progress in the fabrication and application of Ag-MSNs for combination therapies for cancer and infection. We first elaborate on the fabrication of star-shaped structure, core-shell structure, and Janus structure Ag-MSNs. We then highlight Ag-MSNs as a multifunctional nanoplatform to surface-enhanced Raman scattering-based detection, non-photo-based cancer theranostics and photo-based cancer theranostics. In addition, we detail Ag-MSNs for combined antibacterial therapy via drug delivery and phototherapy. Overall, we summarize the challenges and future perspectives of Ag-MSNs that make them promising for diagnosis and therapy of cancer and infection.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Neoplasias , Antibacterianos/química , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Humanos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Dióxido de Silício/química , Prata/química
5.
Small ; 17(26): e2100006, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34081391

RESUMO

Amplifying the chemotherapy-driven immunogenic cell death (ICD) for efficient and safe cancer chemoimmunotherapy remains a challenge. Here, a potential ICD nanoamplifier containing diselenide-bridged mesoporous organosilica nanoparticles (MONs) and chemotherapeutic ruthenium compound (KP1339) to achieve cancer chemoimmunotherapy is tailored. KP1339-loaded MONs show controlled drug release profiles via glutathione (GSH)-responsive competitive coordination and matrix degradation. High concentration of MONs selectively evoked reactive oxygen species production, GSH depletion, and endoplasmic reticulum stress in cancer cells, thus amplifying the ICD of KP1339 and boosting robust antitumor immunological responses. After the combination of PD-L1 checkpoint blockade, cancer cell membrane-cloaked KP1339-loaded MONs not only regress primary tumor growth with low systemic toxicity, but also inhibit distant tumor growth and pulmonary metastasis of breast cancer. The results have shown the potential of coordination and redox dual-responsive MONs boosting amplified ICD for cancer chemoimmunotherapy.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Doxorrubicina , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Morte Celular Imunogênica , Neoplasias/tratamento farmacológico , Oxirredução
6.
Mol Cancer ; 17(1): 14, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29368602

RESUMO

Head and neck cancer (HNC) ranks as the 6th most common malignancy across the world. Metastasis is a hallmark of cancer, primarily contributing to the relapse and poor prognosis of HNC. Recently, long noncoding RNAs (lncRNAs), previously considered as non-functional, are increasingly appreciated by scholars to play crucial roles in mediating HNC metastasis. LncRNAs, which are located in the nucleus and cytoplasm, mainly exert their function via epigenetic modification, transcriptional control and translational regulation. As several lncRNAs are presently demonstrated to participate in HNC metastasis, we make a summary of the functions and mechanisms regarding these lncRNAs. As shown in the literature, most lncRNAs appear to promote the metastasis of HNC. Hence, we primarily discuss the lncRNAs involved in enhancing metastasis. Additionally, more studies are needed to understand those lncRNAs without clear mechanisms. Furthermore, we introduced the upstream regulator for the aberrant expression of lncRNAs in HNC. Finally, we concisely addressed future research prospects of lncRNAs, particularly the interplay between lncRNAs and tumor immunity as well as lncRNA-targeted therapeutic techniques, and we introduced clustered regularly interspaced short palindromic repeats (CRISPR)-Display as a possibly transformative tool to study lncRNAs. Although lncRNA research is still in the initial stage, it holds great promise to be applied as a prognosticator of HNC and a therapeutic target to inhibit HNC metastasis, which could significantly enhance the outcome of HNC patients.


Assuntos
Biomarcadores Tumorais , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , RNA Longo não Codificante/genética , Animais , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/imunologia , Humanos , Metástase Neoplásica , Estadiamento de Neoplasias , Interferência de RNA , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
7.
Ecotoxicol Environ Saf ; 163: 180-187, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30053588

RESUMO

Soil acidification can alter the biogeochemistry of ecosystems and adversely affect biota; however, there are still many debates about the toxicity of aluminum (Al) fractions and Al species in soil:water extracts to plants. In this study, five crude soils with different pH values (4.92-8.51) were collected, seeded with tall fescue and grown in rhizosphere boxes for 120 days. Then, soil properties, labile Al fractions and Al species in soil:water extracts were determined, and their toxicities to plants were analyzed. Our study showed that a stable exchangeable Al fraction (ExAl) pool exists and is supplied by other labile Al fractions. Dissolution of Al from adsorbed hydroxyl-Al fraction (HyAl) and organic-Al fraction (OrAl) may play important roles in soil Al toxicity, as HyAl and OrAl account for major parts of soil labile Al. Additionally, Al3+ and mononuclear hydroxyl-Al species in soil:water extracts have few effects to plants. Nevertheless, high negative correlations were found between Al-F- complexes and tall fescue biomass, indicating their toxicity in the natural soil environment. Thus, in many cases, Al3+ toxicity should not be emphasized because of its lower activity in soil water extracts. Moreover, toxicities of AlF3(aq) and AlF4- to plants should be emphasized, because they have been confirmed in soil water extracts in this study.


Assuntos
Compostos de Alumínio , Alumínio , Festuca/efeitos dos fármacos , Poluentes do Solo , Solo/química , Poluentes da Água , Água/química , Alumínio/efeitos adversos , Alumínio/análise , Compostos de Alumínio/efeitos adversos , Compostos de Alumínio/análise , Biomassa , Festuca/crescimento & desenvolvimento , Poluentes do Solo/efeitos adversos , Poluentes do Solo/análise , Poluentes da Água/efeitos adversos , Poluentes da Água/análise
8.
J Oral Pathol Med ; 46(8): 561-568, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27935123

RESUMO

OBJECTIVES: To evaluate the effectiveness and safety of nucleoside antiviral drugs for the treatment of recurrent herpes labialis. METHODS: Randomized controlled trials that examined the effectiveness and/or safety of nucleoside antiviral drugs for recurrent herpes labialis were identified via a literature search. The parameters used to measure efficacy were time to healing of classic and all lesions, time to resolution of pain, and percentage of aborted lesions. Safety was assessed by evaluating the adverse events reported during treatment. Subgroup analyses based on the mode of application (topical/systemic) and type of nucleoside antiviral drugs were performed, as were sensitivity analyses of studies with a low risk of bias. RESULTS: Our analysis included 16 publications reporting 25 randomized controlled trials (8453 patients). Nucleoside antiviral drugs decreased the time to healing of all lesions (mean difference: -0.74 days; 95% confidence interval: -0.86, -0.62), especially classic lesions (mean difference: -1.09 days; 95% confidence interval: -1.27, -0.92). They also reduced the time to resolution of pain (mean difference: -0.38 days; 95% confidence interval: -0.58, -0.18) and increased the percentage of aborted lesions (rate ratio: 1.15; 95% confidence interval: 1.07, 1.23). Valaciclovir more effectively reduced the time to healing of all lesions and the time to resolution of pain than did aciclovir. Both nucleoside antiviral drugs increased the percentage of aborted lesions, whereas penciclovir and famciclovir did not. CONCLUSIONS: Nucleoside antiviral drugs are safe and beneficial for the treatment of recurrent herpes labialis; both systemic and topical formulations are recommended. Valaciclovir is more effective than aciclovir, especially in reducing the time to healing of lesions.


Assuntos
Antivirais/uso terapêutico , Herpes Labial/tratamento farmacológico , Nucleosídeos/uso terapêutico , Simplexvirus/efeitos dos fármacos , Antivirais/efeitos adversos , Humanos , Nucleosídeos/efeitos adversos , Recidiva
9.
J Oral Pathol Med ; 46(3): 201-207, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27465776

RESUMO

BACKGROUND: 5-Fluorouracil (5-FU) is an essential chemotherapeutic agent for oral squamous cell carcinoma (OSCC). However, toxic side effects have limited its role in OSCC therapy. The aim of this study was to explore whether combination therapy with 5-FU and honokiol (HNK), a small natural organic molecule shown to induce apoptosis in OSCC cells, could enhance the anticancer activity of 5-FU without notably increasing its toxicity. METHODS: 5-FU and/or HNK were used to treat OSCC cells both in vitro and in vivo. The therapeutic effect and underlying mechanisms were evaluated by cell viability assay, flow cytometry, OSCC xenograft mouse model, and Western blot. Tumor tissue apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay. Toxicity was assessed following hematoxylin and eosin staining. RESULTS: Exposure to HNK + 5-FU produced a synergistic cytotoxic effect on OSCC cells. Both HNK and 5-FU could induce apoptosis through the mitochondria-mediated intrinsic pathway, and their specific signaling pathways were different. In the mouse OSCC xenograft model, treatment with 5-FU + HNK substantively retarded tumor growth, as compared to treatment with either drug individually. TUNEL analysis further confirmed that the superior in vivo antitumor efficacy of 5-FU + HNK was associated with enhanced stimulation of cell apoptosis. Notably, HNK did not increase the toxicity of 5-FU. CONCLUSION: These findings suggest that HNK and 5-FU exert a synergistic therapeutic effect on OSCC by inducing apoptosis. HNK might thus enhance the clinical therapeutic efficacy of 5-FU without increasing its toxicity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Fluoruracila/farmacologia , Lignanas/farmacologia , Neoplasias Bucais/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Fluoruracila/uso terapêutico , Humanos , Lignanas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Bucais/patologia
11.
J Periodontol ; 95(2): 175-184, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37469140

RESUMO

BACKGROUND: The association between periodontitis and cardiovascular disease (CVD) has been widely explored, but little is known about the effect of periodontitis on the mortality of CVD patients. This study aims to clarify the effect of periodontitis on all-cause and cause-specific mortality of CVD patients. METHODS: We included 2,135 individuals with CVD from the National Health and Nutrition Examination Survey. Mortality data were ascertained by linkage to National Death Index records through 31 December 2019. We used Cox proportional hazards models for all-cause mortality and competing risk models for CVD and cancer mortality to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Further covariate adjustments, stratification analyses, and a variety of sensitivity analyses were conducted to test the reliability and robustness of the results. RESULTS: The all-cause mortality in CVD patients with moderate/severe periodontitis was significantly higher than in those with no/mild periodontitis (HR: 1.25; 95% CI: 1.02-1.52; P = 0.03). The all-cause mortality in participants with severe clinical attachment loss was significantly higher (HR: 1.07; 95% CI: 1.01-1.14; P = 0.01). However, no discrepancy in CVD or cancer mortality was observed between CVD patients with different periodontal status. CONCLUSIONS: Our findings from a longitudinal study with a large sample indicated significant but slightly higher all-cause mortality in CVD patients with moderate/severe periodontitis than in those with no/mild periodontitis.


Assuntos
Doenças Cardiovasculares , Neoplasias , Periodontite , Humanos , Estudos de Coortes , Estudos Longitudinais , Inquéritos Nutricionais , Doenças Cardiovasculares/complicações , Reprodutibilidade dos Testes , Periodontite/complicações
12.
Adv Mater ; : e2402966, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39044607

RESUMO

Despite the promise of activatable chemotherapy, the development of a spatiotemporally controllable strategy for prodrug activation in deep tissues remains challenging. Herein, a proof-of-concept is proposed for a gold nanocluster-based strategy that utilizes X-ray irradiation to trigger the liberation of platinum (Pt)-based prodrug conjugates, thus enabling radiotherapy-directed chemotherapy. Mechanistically, the irradiated activation of prodrugs is achieved through direct photoelectron transfer from the excited-state gold nanoclusters to the Pt(IV) center, resulting in the release of cytotoxic Pt(II) agents. Compared to the traditional combination of chemotherapy and radiotherapy, this radiotherapy-directed chemotherapy strategy offers superior antitumor efficacy and safety benefits through spatiotemporal synergy at the tumor site. Additionally, this strategy elicits robust immunogenic cell death and yields profound outcomes for combined immunotherapy of breast cancer. This versatile strategy is ushering in a new era of radiation-mediated chemistry for controlled drug delivery and the precise regulation of biological processes.

13.
Biomaterials ; 305: 122465, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38190768

RESUMO

Bacterial infection and delayed osseointegration are two major challenges for titanium-based orthopedic implants. In the present study, we developed a functionalized titanium implant Ti-M@A by immobilizing antimicrobial peptide (AMP) HHC36-loaded diselenide-bridged mesoporous silica nanoparticles (MSNs) on the surface, which showed good long-term and mechanical stability. The functionalized implants can realize the sustained release of AMP over 30 days and exhibit over 95.71 % antimicrobial activity against four types of clinical bacteria (S. aureus, E. coli, P. aeruginosa and MRSA), which arose from the capability to destroy the bacterial membranes. Moreover, Ti-M@A can efficiently inhibit the biofilm formation of the bacteria. The functionalized implants can also significantly promote the osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (mBMSCs) because of the Se in MSNs. Notably, it can trigger macrophages toward M2 polarization in vitro by scavenging ROS in LPS-activated macrophages. Consequently, in vivo assays with infection and non-infection bone defect models demonstrated that such bioactive implants can not only kill over 98.82 % of S. aureus, but also promote osseointegration. Hence, this study provides a combined strategy to resolve bacterial infection and delayed osseointegration for titanium implants.


Assuntos
Infecções Bacterianas , Nanopartículas , Camundongos , Animais , Osseointegração , Titânio/farmacologia , Staphylococcus aureus , Osteogênese , Dióxido de Silício , Escherichia coli , Bactérias , Inflamação , Peptídeos Antimicrobianos , Propriedades de Superfície
14.
Adv Sci (Weinh) ; 11(30): e2400630, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38867377

RESUMO

Senescent cancer cells are endowed with high immunogenic potential that has been leveraged to elicit antitumor immunity and potentially complement anticancer therapies. However, the efficacy of live senescent cancer cell-based vaccination is limited by interference from immunosuppressive senescence-associated secretory phenotype and pro-tumorigenic capacity of senescent cells. Here, a senescent cancer cell-based nanovaccine with strong immunogenicity and favorable potential for immunotherapy is reported. The biomimetic nanovaccine integrating a senescent cancer cell membrane-coated nanoadjuvant outperforms living senescent cancer cells in enhancing dendritic cells (DCs) internalization, improving lymph node targeting, and enhancing immune responses. In contrast to nanovaccines generated from immunogenic cell death-induced tumor cells, senescent nanovaccines facilitate DC maturation, eliciting superior antitumor protection and improving therapeutic outcomes in melanoma-challenged mice with fewer side effects when combined with αPD-1. The study suggests a versatile biomanufacturing approach to maximize immunogenic potential and minimize adverse effects of senescent cancer cell-based vaccination and advances the design of biomimetic nanovaccines for cancer immunotherapy.


Assuntos
Vacinas Anticâncer , Senescência Celular , Imunoterapia , Animais , Camundongos , Vacinas Anticâncer/imunologia , Imunoterapia/métodos , Senescência Celular/imunologia , Modelos Animais de Doenças , Biomimética/métodos , Camundongos Endogâmicos C57BL , Membrana Celular/imunologia , Nanopartículas , Humanos , Linhagem Celular Tumoral , Feminino , Materiais Biomiméticos , Células Dendríticas/imunologia , Nanovacinas
15.
Adv Sci (Weinh) ; 11(10): e2308866, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38196299

RESUMO

Exploration of medicines for efficient and safe management of metabolic-associated fatty liver disease (MAFLD) remains a challenge. Obeticholic acid (OCA), a selective farnesoid X receptor agonist, has been reported to ameliorate injury and inflammation in various liver diseases. However, its clinical application is mainly limited by poor solubility, low bioavailability, and potential side effects. Herein a hepatic-targeted nanodrugs composed of OCA and cholesterol-lowering atorvastatin (AHT) with an ideal active pharmaceutical ingredient (API) content for orally combined treatment of MAFLD is created. Such carrier-free nanocrystals (OCAHTs) are self-assembled, not only improving the stability in gastroenteric environments but also achieving hepatic accumulation through the bile acid transporter-mediated enterohepatic recycling process. Orally administrated OCAHT outperforms the simple combination of OCA and AHT in ameliorating of liver damage and inflammation in both acetaminophen-challenged mice and high-fat diet-induced MAFLD mice with less systematic toxicity. Importantly, OCAHT exerts profoundly reverse effects on MAFLD-associated molecular pathways, including impairing lipid metabolism, reducing inflammation, and enhancing the antioxidation response. This work not only provides a facile bile acid transporter-based strategy for hepatic-targeting drug delivery but also presents an efficient and safe full-API nanocrystal with which to facilitate the practical translation of nanomedicines against MAFLD.


Assuntos
Ácido Quenodesoxicólico/análogos & derivados , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Atorvastatina/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Inflamação/tratamento farmacológico
16.
Acta Biomater ; 174: 281-296, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37951519

RESUMO

RNA interference (RNAi) presents great potential against intractable liver diseases. However, the establishment of specific, efficient, and safe delivery systems targeting hepatocytes remains a great challenge. Herein, we described a promising hepatocytes-targeting system through integrating triantennary N-acetylgalactosamine (GalNAc)-engineered cell membrane with biodegradable mesoporous silica nanoparticles, which efficiently and safely delivered siRNA to hepatocytes and silenced the target PCSK9 gene expression for the treatment of non-alcoholic fatty liver disease. Having optimized the GalNAc-engineering strategy, insertion orders, and cell membrane source, we obtained the best-performing GalNAc-formulations allowing strong hepatocyte-specific internalization with reduced Kupffer cell capture, resulting in robust gene silencing and less hepatotoxicity when compared with cationic lipid-based GalNAc-formulations. Consequently, a durable reduction of lipid accumulation and damage was achieved by systemic administering siRNAs targeting PCSK9 in high-fat diet-fed mice, accompanied by displaying desirable safety profiles. Taken together, this GalNAc-engineering biomimetics represented versatile, efficient, and safe carriers for the development of hepatocyte-specific gene therapeutics, and prevention of metabolic diseases. STATEMENT OF SIGNIFICANCE: Compared to MSN@LP-GN3 (MC3-LNP), MSN@CM-GN3 exhibited strong hepatocyte targeting and Kupffer cell escaping, as well as good biocompatibility for safe and efficient siRNA delivery. Furthermore, siPCSK9 delivered by MSN@CM-GN3 reduced both serum and liver LDL-C, TG, TC levels and lipid droplets in HFD-induced mice, resulting in better performance than MSN/siPCSK9@LP-GN3 in terms of lipid-lowering effect and safety profiles. These findings indicated promising advantages of our biomimetic GN3-based systems for hepatocyte-specific gene delivery in chronic liver diseases. Our work addressed the challenges associated with the lower targeting efficiency of cell membrane-mimetic drug delivery systems and the immunogenicity of traditional GalNAc delivery systems. In conclusion, this study provided an effective and versatile approach for efficient and safe gene editing using ligand-integrated biomimetic nanoplatforms.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Pró-Proteína Convertase 9 , Camundongos , Animais , Interferência de RNA , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/farmacologia , Hepatopatia Gordurosa não Alcoólica/terapia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Biomimética , Hepatócitos/metabolismo , Fígado/metabolismo , RNA Interferente Pequeno/farmacologia , Lipídeos/farmacologia
17.
Adv Mater ; 36(32): e2314197, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38713519

RESUMO

Combining radiotherapy with immune checkpoint blockade therapy offers a promising approach to treat glioblastoma multiforme (GBM), yet challenges such as limited effectiveness and immune-related adverse events (irAEs) persist. These issues are largely due to the failure in targeting immunomodulators directly to the tumor microenvironment. To address this, a biomimetic nanoplatform that combines a genetically modified mesenchymal stem cell (MSC) membrane with a bioactive nanoparticle core for chemokine-directed radioimmunotherapy of GBM is developed. The CC chemokine receptor 2 (CCR2)-overexpressing MSC membrane acts as a tactical tentacle to achieve radiation-induced tropism toward the abundant chemokine (CC motif) ligand 2 (CCL2) in irradiated gliomas. The nanoparticle core, comprising diselenide-bridged mesoporous silica nanoparticles (MSNs) and PD-L1 antibodies (αPD-L1), enables X-ray-responsive drug release and radiosensitization. In two murine models with orthotopic GBM tumors, this nanoplatform reinvigorated immunogenic cell death, and augmented the efficacy and specificity of GBM radioimmunotherapy, with reduced occurrence of irAEs. This study suggests a promising radiation-induced tropism strategy for targeted drug delivery, and presents a potent nanoplatform that enhances the efficacy and safety of radio-immunotherapy.


Assuntos
Glioblastoma , Nanopartículas , Radioimunoterapia , Glioblastoma/radioterapia , Glioblastoma/terapia , Glioblastoma/patologia , Animais , Radioimunoterapia/métodos , Camundongos , Nanopartículas/química , Humanos , Linhagem Celular Tumoral , Células-Tronco Mesenquimais , Antígeno B7-H1/metabolismo , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patologia , Dióxido de Silício/química
18.
ACS Nano ; 18(1): 470-482, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38146673

RESUMO

Targeted delivery of vaccines to the spleen remains a challenge. Inspired by the erythrophagocytotic process in the spleen, we herein report that intravenous administration of senescent erythrocyte-based vaccines profoundly alters their tropism toward splenic antigen-presenting cells (APCs) for imprinting adaptive immune responses. Compared with subcutaneous inoculation, intravenous vaccination significantly upregulated splenic complement expression in vivo and demonstrated synergistic antibody killing in vitro. Consequently, intravenous senescent erythrocyte vaccination produces potent SARS-CoV-2 antibody-neutralizing effects, with potential protective immune responses. Moreover, the proposed senescent erythrocyte can deliver antigens from resected tumors and adjuvants to splenic APCs, thereby inducing a personalized immune reaction against tumor recurrence after surgery. Hence, our findings suggest that senescent erythrocyte-based vaccines can specifically target splenic APCs and evoke adaptive immunity and complement production, broadening the tools for modulating immunity, helping to understand adaptive response mechanisms to senescent erythrocytes better, and developing improved vaccines against cancer and infectious diseases.


Assuntos
Baço , Vacinas , Vacinação , Imunidade Adaptativa , Administração Intravenosa , Eritrócitos
19.
BMJ Case Rep ; 16(12)2023 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-38154872

RESUMO

This case report presents a young girl in her early childhood diagnosed with chronic mucocutaneous candidiasis (CMC) and primary hypothyroidism. Genetic analysis revealed a novel de novo mutation in the STAT1 gene (exon 11, c.972C>G, p.Cys324Trp), adding to the existing literature on STAT1 mutations, which account for approximately 53% of CMC cases. The identified mutation is predicted to have a more severe pathogenic impact based on PolyPhen-2 scoring. Our findings emphasise the importance of comprehensive genetic testing in CMC diagnosis and suggest that the specific mutation site may correlate with disease prognosis. The case underscores the need for vigilant monitoring and targeted therapeutic interventions, given the potential for poorer outcomes.


Assuntos
Candidíase Mucocutânea Crônica , Hipotireoidismo , Feminino , Humanos , Pré-Escolar , Criança , Candidíase Mucocutânea Crônica/diagnóstico , Candidíase Mucocutânea Crônica/genética , Candidíase Mucocutânea Crônica/complicações , Prognóstico , Mutação , Fator de Transcrição STAT1/genética , Testes Genéticos , Hipotireoidismo/complicações , Hipotireoidismo/genética
20.
Chin Med J Pulm Crit Care Med ; 1(2): 102-107, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-39170822

RESUMO

Chronic Obstructive Pulmonary Disease (COPD) is characterized by persistent respiratory symptoms and airflow limitation. Acute exacerbation of COPD (AECOPD) is an acute worsening of respiratory symptoms, which needs additional treatment and can result in worsening health status, increasing risks of hospitalization and mortality. Therefore, it is necessary to early recognize and diagnose exacerbations of COPD. This review introduces the updated definition of COPD exacerbations, the current clinical assessment tools, and the current potential biomarkers. The application of mobile health care in COPD management for early identification and diagnosis is also included in this review.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA