Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39294473

RESUMO

Activation of the Toll-like receptor 4 (TLR4) by bacterial endotoxins in macrophages plays a crucial role in the pathogenesis of sepsis. However, the mechanism underlying TLR4 activation in macrophages is still not fully understood. Here, we reveal that upon lipopolysaccharide (LPS) stimulation, lysine acetyltransferase CBP is recruited to the TLR4 signalosome complex leading to increased acetylation of the TIR domains of the TLR4 signalosome. Acetylation of the TLR4 signalosome TIR domains significantly enhances signaling activation via NF-κB rather than IRF3 pathways. Induction of NF-κB signaling is responsible for gene expression changes leading to M1 macrophage polarization. In sepsis patients, significantly elevated TLR4-TIR acetylation is observed in CD16+ monocytes combined with elevated expression of M1 macrophage markers. Pharmacological inhibition of HDAC1, which deacetylates the TIR domains, or CBP play opposite roles in sepsis. Our findings highlight the important role of TLR4-TIR domain acetylation in the regulation of the immune responses in sepsis, and we propose this reversible acetylation of TLR4 signalosomes as a potential therapeutic target for M1 macrophages during the progression of sepsis.

2.
Trends Immunol ; 44(12): 1031-1045, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37932176

RESUMO

Tumor immunotherapy is refashioning traditional treatments in the clinic for certain tumors, especially by relying on the activation of T cells. However, the safety and effectiveness of many antitumor immunotherapeutic agents are suboptimal due to difficulties encountered in assessing T cell responses and adjusting treatment regimens accordingly. Here, we review advances in the clinical visualization of T cell activity in vivo, and focus particularly on molecular imaging probes and biomarkers of T cell activation. Current challenges and prospects are also discussed that aim to achieve a better strategy for real-time monitoring of T cell activity, predicting prognoses and responses to tumor immunotherapy, and assessing disease management.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Linfócitos T , Neoplasias/terapia , Imunoterapia/métodos , Imagem Molecular
3.
J Am Chem Soc ; 146(32): 22455-22468, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39094119

RESUMO

Myeloid-derived suppressor cells (MDSCs) significantly hinder the immune response to tumor radiotherapy (RT) because of their massive accumulation in tumors after RT, resulting in immunosuppression and poor clinical prognosis. Herein, we developed an anti-PD-L1 antibody-conjugated iron oxide nanoprobe (Fe3O4-αPD-L1) to target and induce ferroptosis in MDSCs, thereby alleviating RT resistance. Overexpression of PD-L1 in MDSCs following RT enables noninvasive in vivo magnetic resonance and positron emission tomography imaging using 89Zr-labeled nanoprobes to track the movement of MDSCs and their infiltration into the tumor. After uptake by MDSCs that infiltrated the tumor, Fe3O4-αPD-L1 nanoprobes were mainly found within the lysosome and triggered the Fenton reaction, resulting in the generation of abundant reactive oxygen species. This process leads to ferroptosis of MDSCs, characterized by lipid peroxidation and mitochondrial dysfunction, and effectively reprograms the immunosuppressive environment within the tumor following RT. This study highlights a strategy for monitoring and regulating the fate of MDSCs to alleviate RT resistance and ultimately achieve improved treatment outcomes.


Assuntos
Ferroptose , Células Supressoras Mieloides , Ferroptose/efeitos dos fármacos , Células Supressoras Mieloides/imunologia , Animais , Humanos , Camundongos , Antígeno B7-H1/metabolismo , Neoplasias/diagnóstico por imagem , Neoplasias/radioterapia , Neoplasias/patologia , Compostos Férricos/química , Linhagem Celular Tumoral
4.
Crit Rev Food Sci Nutr ; : 1-20, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-38062765

RESUMO

Epigenetics regulates gene expression and play significant roles across diverse disease states. Epigenetics mechanisms, including DNA methylation, histone modifications, microRNAs/lncRNA, and N6-methyladenosine (m6A) RNA methylation, elicit heritable but reversible modifications in gene expression without modifying the DNA sequence. Recent research suggests that certain natural phytochemicals with chemopreventive properties have the potential to function as epigenetic regulators. Quercetin, a derivative of natural flavonoid glycosides and a constituent of the human diet, is linked to a variety of health benefits including anti-inflammatory, anticancer activity, antiapoptotic, antihypertensive, and neuroprotective effects. Recent findings suggest that quercetin possesses the ability to modulate canonical biochemical signaling pathways and exert an impact on epigenetic networks. This review aims to synthesize the most recent research findings that elucidate the potential biological effects of quercetin and its influence on in vitro and in vivo models via epigenetic mechanisms. In light of our findings, it is evident that quercetin possesses the potential to function as an exemplary instance of naturally derived phytochemicals, which can be effectively employed as a pivotal constituent in functional foods and dietary supplements aimed at the amelioration of various ailments. More specifically, its mechanism of action involves the alteration of diverse epigenetic targets.

5.
Analyst ; 148(24): 6334-6340, 2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-37947486

RESUMO

Indocyanine green (ICG), as the only Federal Drug Administration (FDA) approved fluorescence imaging agent, has been widely applied in clinics for near-infrared (NIR) fluorescence imaging-guided surgery and photothermal therapy of cancers. However, its lack of target specificity and poor photo and photothermal stabilities seriously restrict its wide application in clinical practice. Herein, we developed ICG-derived NIR fluorescent probes consisting of a cypate fluorophore and one or two cyclic-(arginine-glycine-aspartic acid) (cRGD) peptides that can specifically target αvß3 integrin for accurate diagnosis and therapy of oral tumors. Probe Cy-2RGD has been demonstrated to possess bright NIR emission, great tumor targeting capability and a photothermal effect. Moreover, it could be successfully used for effective imaging-guided surgical resection as well as photothermal therapy of oral tumors. This work could provide a valuable tool for sensitive detection and accurate treatment of malignant tumors.


Assuntos
Verde de Indocianina , Neoplasias Bucais , Humanos , Verde de Indocianina/uso terapêutico , Terapia Fototérmica , Corantes Fluorescentes , Neoplasias Bucais/diagnóstico por imagem , Neoplasias Bucais/terapia , Imagem Óptica/métodos
6.
Immunology ; 167(4): 471-481, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36065492

RESUMO

The immune checkpoint programmed death receptor 1 (PD-1) and programmed death ligand 1 (PD-L1) are biologically important immunosuppressive molecules, and the PD-L1/PD-1-mediated signalling pathway is currently considered one of the main mechanisms of tumour escape immune surveillance. PD-L1 is highly expressed on the cytomembrane of tumour cell and binds to PD-1 receptor of activated T cells. This interaction activates PD-L1/PD-1 downstream signal transduction, inhibiting T cells anti-tumour activity. Therefore, inhibitors of PD-L1/PD-1 activation, showing significant efficacy in some types of tumours, have been widely approved in clinical tumour therapy. Recent research on PD-L1/PD-1 signalling pathway regulation has shown post-translational modifications (PTMs) form of PD-L1 or PD-1, including glycosylation, ubiquitination, phosphorylation, and acetylation, which may play an important role in PD-L1/PD-1 signalling pathway regulation and anti-tumour function of T cells. In this review, we focused on PTMs of PD-L1/PD-1 research and potential applications in tumour immunotherapy.


Assuntos
Antígeno B7-H1 , Neoplasias , Humanos , Receptor de Morte Celular Programada 1 , Imunoterapia , Processamento de Proteína Pós-Traducional
7.
Anal Chem ; 93(26): 9277-9285, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34160212

RESUMO

Specific and effective accumulation of nanoparticles within tumors is highly crucial for precise cancer diagnosis and treatment. Therefore, spatiotemporally manipulating the aggregation of small gold nanoparticles (AuNPs) in a tumor microenvironment is of great significance for enhancing the diagnostic and therapeutic efficacy of tumors. Herein, we reported a novel furin enzyme/acidic pH synergistically triggered small AuNP aggregation strategy for activating the photoacoustic (PA) imaging and photothermal (PTT) functions of AuNPs in vivo. Smart gold nanoparticles decorated with furin-cleavable RVRR (Arg-Val-Arg-Arg) peptides (Au-RRVR) were rationally designed and fabricated. Both in vitro and in vivo experiments demonstrated that such Au-RRVR nanoparticles could be simultaneously induced by furin and acidic pH to form large aggregates within tumorous tissue resulting in improved tumor accumulation and retention, which can further activate the PA and PTT effect of AuNPs for sensitive imaging and efficient therapy of tumors. Thus, we believe that this dual-stimuli-responsive aggregation system may offer a universal platform for effective cancer diagnosis and treatment.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Linhagem Celular Tumoral , Furina , Ouro , Humanos , Concentração de Íons de Hidrogênio , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Fototerapia , Terapia Fototérmica , Microambiente Tumoral
8.
J Am Chem Soc ; 142(51): 21502-21512, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33306393

RESUMO

Improving the enrichment of drugs or theranostic agents within tumors is very vital to achieve effective cancer diagnosis and therapy while greatly reducing the dosage and damage to normal tissues. Herein, as a proof of concept, we for the first time report a red light-initiated probe-RNA cross-linking (RLIPRC) strategy that can not only robustly promote the accumulation and retention of the probe in the tumor for prolonged imaging but also significantly inhibits the tumor growth. A near-infrared (NIR) fluorescent probe f-CR consisting of a NIR dye (Cyanine 7) as a signal reporter, a cyclic-(arginine-glycine-aspartic acid) (cRGD) peptide for tumor targeting, and a singlet oxygen (1O2)-sensitive furan moiety for RNA cross-linking was rationally designed and synthesized. This probe possessed both passive and active tumor targeting abilities and emitted intense NIR/photoacoustic (PA) signals, allowing for specific and sensitive dual-modality imaging of tumors in vivo. Notably, probe f-CR could be specifically and covalently cross-linked to cytoplasmic RNAs via the cycloaddition reaction between furan and adenine, cytosine, or guanine under the oxidation of 1O2 generated in situ by irradiation of methylene blue (MB) with 660 nm laser light, which effectively blocks the exocytosis of the probes resulting in enhanced tumor accumulation and retention. More excitingly, for the first time, we revealed that the covalent cross-linking of probe f-CR to cytoplasmic RNAs could induce severe apoptosis of cancer cells leading to remarkable tumor suppression. This study thus represents the first red light-initiated RNA cross-linking system with high potential to improve the diagnostic and therapeutic outcomes of tumors in vivo.


Assuntos
Citoplasma/metabolismo , Raios Infravermelhos , Imagem Molecular/métodos , Fotoquimioterapia/métodos , RNA/metabolismo , Carbocianinas/química , Linhagem Celular Tumoral , Citoplasma/efeitos da radiação , Humanos , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo
9.
Chembiochem ; 20(5): 667-671, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30447100

RESUMO

Manipulating the cross-coupling of gold nanoparticles (AuNPs) to maximize the photothermal effect is a promising strategy for cancer therapy. Here, by taking advantage of the well-known tetrazole/alkene photoclick chemistry, we have demonstrated for the first time that small AuNPs (23 nm) decorated with both 2,5-diphenyltetrazole and methacrylic acid on their surfaces can form covalently crosslinked aggregates upon laser irradiation (λ=405 nm). In vitro studies indicated that the light-triggered assembling shifted the surface plasmon resonance of AuNPs significantly to near-infrared (NIR) regions, which as a consequence effectively enhanced the efficacy of photothermal therapy for 4T1 breast cancer cells. We thus believe that this new light-triggered cross-coupling approach might offer a valuable tool for cancer treatment.


Assuntos
Ouro/uso terapêutico , Hipertermia Induzida/métodos , Nanopartículas Metálicas/uso terapêutico , Neoplasias/tratamento farmacológico , Fototerapia/métodos , Células 3T3 , Animais , Linhagem Celular Tumoral , Metacrilatos/química , Camundongos , Ressonância de Plasmônio de Superfície , Tetrazóis/química
10.
Chemistry ; 23(56): 13893-13896, 2017 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-28884843

RESUMO

A simple and mild light-triggered fluorescent labeling approach, based on the photolysis and reactive carbene insertion of diazirine, to rapidly generate fluorescent nanoparticles is reported. Proof of concept studies demonstrated that the non-fluorescent SiNPs could be covalently labeled by diazirine-conjugated fluorescein (dFITC) upon photo irradiation (λ=365 nm) to afford fluorescent SiNPs with good fluorescent stability and low cytotoxicity. In vitro cellular imaging results indicated that the internalization of SiNPs into living cells could be readily visualized based on fluorescein-labeled SiNPs (F-SiNPs) in real time. We believe that the light-triggered labeling approach may offer a valuable method for surface modification of nanomaterials to achieve new functions.

11.
J Control Release ; 365: 398-411, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38007194

RESUMO

Secretory otitis media (SOM) is a clinical condition characterized by the accumulation of fluids and oxidative stress in the middle ear, leading to hearing impairment and infection complications. One potential solution for mitigating oxidative stress associated with SOM is the use of antioxidants such as astaxanthin. However, its effectiveness is limited due to its poor bioavailability and rapid oxidation. Herein, we developed a novel diselenium-crosslinked apotransferrin enriched with astaxanthin (AST@dSe-AFT) nanoparticles to augment the transport of astaxanthin across biological membranes, resulting in increased bioavailability and reduced oxidative stress in SOM. Our research demonstrated that AST@dSe-AFT efficiently accumulated in the middle ear, allowing for controlled delivery of astaxanthin in response to reactive oxygen species and reducing oxidative stress. Additionally, AST@dSe-AFT stimulated macrophages to polarize towards M2 phenotype and neutrophils to polarize towards N2 phenotype, thereby facilitating an anti-inflammatory response and tissue restoration. Importantly, AST@dSe-AFT exhibited no toxicity or adverse effects, suggesting its potential for safety and future clinical translation. Our findings suggested that AST@dSe-AFT represents a promising approach for the treatment of secretory otitis media and other oxidative stress-related disorders.


Assuntos
Apoproteínas , Nanopartículas , Otite Média com Derrame , Transferrina , Humanos , Otite Média com Derrame/tratamento farmacológico , Antioxidantes/uso terapêutico , Estresse Oxidativo , Xantofilas
12.
Biomater Adv ; 158: 213764, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38227991

RESUMO

Neutrophils play a crucial role in inflammatory immune responses, but their in vivo homing to inflammatory lesions remains unclear, hampering precise treatment options. In this study, we employed a biomineralization-inspired multimodal nanoagent to label neutrophils, enabling noninvasive monitoring of the dynamic process of inflammatory recruitment and guiding photothermal therapy in rheumatoid arthritis. Our nanoagents allowed visualization of neutrophil fate through magnetic resonance imaging, photoacoustic imaging, and fluorescence imaging in the first and second near-infrared windows. Histopathology and immunofluorescence analysis revealed pronounced inflammatory cell infiltration in rheumatoid arthritis compared to the normal limb. Furthermore, the recruitment quantity of neutrophils positively correlated with the inflammatory stage. Additionally, the inherent photothermal effect of the nanoagents efficiently ablated inflammatory cells during the optimal homing time and inflammatory phase. This neutrophil imaging-guided photothermal therapy precisely targeted inflammatory nuclei in rheumatoid arthritis and downregulated pro-inflammatory cytokines in serum. These results demonstrate that in vivo tracking of inflammatory immune response cells can significantly optimize the treatment of inflammatory diseases, including rheumatoid arthritis.


Assuntos
Artrite Reumatoide , Neutrófilos , Humanos , Fototerapia , Terapia Fototérmica , Artrite Reumatoide/terapia , Biomineralização
13.
ACS Nano ; 18(1): 1022-1040, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38131289

RESUMO

In situ vaccination (ISV) formed with the aid of intratumorally injected adjuvants has shed bright light on enhancing the abscopal therapeutic effects of radiotherapy. However, the limited availability of antigens resulting from the radiotherapy-induced immunogenic cell death largely hampers the clinical outcome of ISV. To maximally utilize the radiotherapy-induced antigen, we herein developed a strategy by capturing the radiotherapy-induced antigen in situ with a nanoadjuvant comprised of CpG-loaded Fe3O4 nanoparticles. The highly efficient click reaction between the maleimide residue on the nanoadjuvant and sulfhydryl group on the antigen maximized the bioavailability of autoantigens and CpG adjuvant in vivo. Importantly, combined immune checkpoint blockade can reverse T cell exhaustion after treatment with radiotherapy-induced ISV, thereby largely suppressing the treated and distant tumor. Mechanistically, metabolomics reveals the intratumorally injected nanoadjuvants disrupt redox homeostasis in the tumor microenvironment, further inducing tumor ferroptosis after radiotherapy. Overall, the current study highlights the immense potential of the innovative antigen-capturing nanoadjuvants for synergistically enhancing the antitumor effect.


Assuntos
Imunoterapia , Neoplasias , Humanos , Imunoterapia/métodos , Neoplasias/radioterapia , Imunidade , Adjuvantes Imunológicos/farmacologia , Vacinação , Microambiente Tumoral , Linhagem Celular Tumoral
14.
ACS Nano ; 18(34): 23741-23756, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39158207

RESUMO

Cell backpacks present significant potential in both therapeutic and diagnostic applications, making it essential to further explore their interactions with host cells. Current evidence indicates that backpacks can induce sustained immune responses. Our original objective was to incorporate a model antigen into the backpacks to promote dendritic cell maturation and facilitate antigen presentation, thereby inducing immune responses. However, we unexpectedly discovered that both antigen-loaded backpacks and empty backpacks demonstrated comparable abilities to induce dendritic cell maturation, resulting in nearly identical potency in T-cell proliferation. Our mechanistic studies suggest that the attachment of backpacks induces mechanical forces on dendritic cells via opening the PIEZO1 mechanical ion channel. This interaction leads to the remodeling of the intracellular cytoskeleton and facilitates the production of type I interferons by dendritic cells. Consequently, the mechano-immune-driven dendritic cell backpacks, when combined with radiotherapy, induce a robust antitumor effect. This research presents an avenue for leveraging mechanotransduction to enhance combination immunotherapeutic strategies, potentially leading to groundbreaking advancements in the field.


Assuntos
Células Dendríticas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Camundongos , Mecanotransdução Celular/imunologia , Camundongos Endogâmicos C57BL , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/patologia , Neoplasias/radioterapia , Proliferação de Células/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
15.
ACS Nano ; 18(28): 18425-18443, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38975713

RESUMO

Tumor in situ vaccination (ISV) strategies have emerged in clinical trials as promising approaches, involving the release of tumor antigens through local radiotherapy and intratumorally adjuvant injections. However, the current fabrication strategy for achieving a sustainable immune response to ISV remains a pressing challenge. In this study, we present an empowered sustainable ISV method for antitumor therapy using 177Lu-labeled manganese-doped mesoporous hydroxyapatite (177Lu/Mn-HAP) microspheres. The ISV enables the sustained utilization of tumor antigens, leading to the activation of dendritic cells and polarization of macrophages toward the M1 subtype. Consequently, it facilitates the generation of potent CD8+ T-cell responses, enhancing the antitumor effects of internal radiation in both primary and distant tumors. Importantly, this approach achieves complete remission in all tumor-bearing mice and stimulates immune memory to prevent tumor recurrence. Our study highlights a universal and safe ISV strategy capable of inducing potent tumor-specific and sustainable immune response.


Assuntos
Vacinas Anticâncer , Durapatita , Microesferas , Durapatita/química , Animais , Camundongos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/química , Linfócitos T CD8-Positivos/imunologia , Vacinação , Feminino , Camundongos Endogâmicos C57BL , Radioisótopos/química , Linhagem Celular Tumoral
16.
ACS Nano ; 18(35): 23941-23957, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39171773

RESUMO

Cuproptosis is an emerging form of cell death that relies on the targeted delivery of copper ions to lipoylated tricarboxylic acid cycle proteins. However, a major challenge associated with cuproptosis is its potential to kill both normal and tumor cells without discrimination. Therefore, it is crucial to develop strategies for precise intracellular delivery and redox control of copper to create effective cuproptosis-based tumor therapies. We have introduced a class of nanoagents called metabolism aiming Cu2-xS (MACuS) through a glucose-mediated biomineralization approach. MACuS nanoagents can be specifically targeted to tumors via the glucose transport receptor 1, and we found that NIR-II irradiation can not only result in direct hyperthermia ablation of tumor cells but also facilitate efficient cuproptosis and enhance reactive oxygen species-induced cytotoxicity in tumor cells. As a result, the triple effect of MACuS treatment induced immunogenic cell death, which triggered systemic antitumor immune responses and demonstrated potent efficacy in inhibiting growth, metastasis, and recurrence in mouse and rabbit breast cancer models. The precise intracellular delivery and redox control of copper provided by MACuS hold great potential for the development of highly efficient cuproptosis-based tumor therapies with minimal off-target effects.


Assuntos
Cobre , Animais , Cobre/química , Cobre/farmacologia , Camundongos , Coelhos , Feminino , Humanos , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/química , Camundongos Endogâmicos BALB C , Terapia Fototérmica , Proliferação de Células/efeitos dos fármacos
17.
ACS Nano ; 18(4): 2800-2814, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38227969

RESUMO

The development of a radioresponsive delivery platform has led to an innovative combination radioimmunotherapy strategy for treating tumors. However, controlling the release of immunomodulators by local radiotherapy in vivo remains a significant challenge in order to minimize off-target toxicity, reduce radiation-induced immunosuppression, and maximize synergistic radioimmunotherapy efficacy. In this study, we report the development of core-cross-linked diselenide nanoparticles (dSeNPs) as carriers for radioresponsive delivery of the toll-like receptors 7/8 agonist through systemic administration to achieve combined radioimmunotherapy of tumors. The dSeNPs were fabricated from a ring-opening reaction between 2,2'-diselenidebis(ethylamine) and the ethylene oxide group of an amphiphilic block copolymer. The diselenide bonds were naturally protected in the core of the self-assembled nanostructure, making the dSeNPs extremely stable in the physiological environment. However, they exhibited dose- and time-dependent radiosensitivity, meaning that X-ray irradiation could spatiotemporally control the release of R848 from the dSeNPs. In vivo results showed that local radioresponsive R848 release from dSeNPs greatly improved the synergistic efficacy of combined radioimmunotherapy via the programmed cooperative immune system activation process. This process included macrophage polarization, dendritic cell maturation, and cytotoxic T cell activation. Our findings suggest that core-cross-linked dSeNPs are a promising platform for combined radiotherapy due to their spatiotemporal controllability of radioresponsive drug release.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Receptor 7 Toll-Like/agonistas , Radioimunoterapia , Neoplasias/tratamento farmacológico , Adjuvantes Imunológicos , Nanopartículas/química
18.
Chem Biol Interact ; 403: 111256, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39332791

RESUMO

Bismuth compounds, particularly colloidal bismuth subcitrate (CBS), have been widely used in the treatment of gastrointestinal diseases. However, overdose of CBS has been linked to cases of acute renal failure, primarily due to the intracellular accumulation of bismuth in the kidney. To date, the detailed mechanisms of CBS internalization and its metabolic fate remain unclear. In this study, CBS was characterized as a type of nano-object using transmission electron microscopy and dynamic light scattering. Renal cells internalized CBS primarily via clathrin-mediated endocytosis in an active transport manner. Gene knockdown techniques revealed that CBS binds to the transferrin receptor likely through complexing with transferrin before cellular uptake. Once internalized, CBS was sorted into early endosomes, late endosomes, and lysosomes, mediated by microtubules and the Golgi apparatus. Additionally, differentially expressed genes analysis revealed that CBS endocytosis stimulated oxidative stress, significantly affecting the metabolism of glutathione and cysteine within cells. This led to the formation of black bismuth sulfide particles as a result of CBS conjugating with intracellular glutathione. These findings provide crucial insights into the cellular mechanisms underlying excessive CBS exposure, which is essential for understanding and potentially mitigating the risks associated with the use of bismuth compounds in medical treatments.

19.
ACS Nano ; 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39066710

RESUMO

Radiotherapy (RT)-induced in situ vaccination greatly promotes the development of personalized cancer vaccines owing to the massive release of antigens initiated by tumor-localized RT eliciting the tumor-specific immune response. However, its broad application in cancer treatment is seriously impeded by poor antigen cross-presentation, low response rate, and short duration of efficacy. Herein, the tumor-antigen-capturing nanosystem dAuNPs@CpG consisting of gold nanoparticles, 3,5-cyclohexanedione (CHD), and immunoadjuvant CpG were fabricated to enhance RT-induced vaccination. Taking advantage of the specific covalent binding between CHD and sulfenic acids of antigen proteins, we show that this nanoplatform has an unexpected potential to capture the sulfenylated tumor-derived protein antigens (TDPAs) induced by RT to in situ generate a vaccination effect, achieving significant growth suppression of both primary and distant tumors in combination with PD-1 blockade. We thus believe that our work presents a powerful and effective means to improve the synergistic tumor radioimmunotherapy.

20.
Chem Sci ; 14(9): 2369-2378, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36873836

RESUMO

Spatiotemporally manipulating the in situ immobilization of theranostic agents within cancer cells to improve their bioavailability is highly significant yet challenging in tumor diagnosis and treatment. Herein, as a proof-of concept, we for the first time report a tumor-targetable near-infrared (NIR) probe DACF with photoaffinity crosslinking characteristics for enhanced tumor imaging and therapeutic applications. This probe possesses great tumor-targeting capability, intensive NIR/photoacoustic (PA) signals, and a predominant photothermal effect, allowing for sensitive imaging and effective photothermal therapy (PTT) of tumors. Most notably, upon 405 nm laser illumination, DACF could be covalently immobilized within tumor cells through a photocrosslinking reaction between photolabile diazirine groups and surrounding biomolecules resulting in enhanced tumor accumulation and prolonged retention simultaneously, which significantly facilitates the imaging and PTT efficacy of tumor in vivo. We therefore believe that our current approach would provide a new insight for achieving precise cancer theranostics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA