Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 640
Filtrar
1.
J Colloid Interface Sci ; 678(Pt A): 937-949, 2025 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-39226834

RESUMO

Synergistic photothermal/immunotherapy has garnered significant attention for its potential to enhance tumor therapeutic outcomes. However, the fabrication of an intelligent system with a simple composition that simultaneously exerts photothermal/immunotherapy effect and imaging guidance function still remains a challenge. Herein, a glutathione (GSH)-responsive theranostic nanoprobe, named HA-MnO2/ICG, was elaborately constructed by loading photothermal agent (PTA) indocyanine green (ICG) onto the surface of hyaluronic acid (HA)-modified manganese dioxide nanosheets (HA-MnO2) for magnetic resonance (MR) imaging-guided synergetic photothermal/immuno-enhanced therapy. In this strategy, HA-MnO2 nanosheets were triggered by the endogenous GSH in tumor microenvironment to generate Mn2+ for MR imaging, where the longitudinal relaxation rate of HA-MnO2/ICG was up to 14.97 mM-1s-1 (∼24 times than that found in a natural environment), demonstrating excellent intratumoral MR imaging. Moreover, the HA-MnO2/ICG nanoprobe demonstrates remarkable photothermal therapy (PTT) efficacy, generating sufficient heat to induce immunogenic cell death (ICD) within tumor cells. Meanwhile the released Mn2+ ions from the nanosheets function as potent immune adjuvants, amplifying the immune response against cancer. In vivo experiments validated that HA-MnO2/ICG-mediated PTT was highly effective in eradicating primary tumors, while simultaneously enhancing immunogenicity to prevent the growth of distal metastasis. This hybrid HA-MnO2/ICG nanoprobe opened new avenues in the design of MR imaging-monitored PTT/immuno-enhanced synergistic therapy for advanced cancer.


Assuntos
Ácido Hialurônico , Imunoterapia , Verde de Indocianina , Imageamento por Ressonância Magnética , Compostos de Manganês , Óxidos , Terapia Fototérmica , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Animais , Óxidos/química , Camundongos , Ácido Hialurônico/química , Verde de Indocianina/química , Humanos , Antineoplásicos/química , Antineoplásicos/farmacologia , Tamanho da Partícula , Fototerapia , Nanopartículas/química , Propriedades de Superfície , Camundongos Endogâmicos BALB C , Nanomedicina Teranóstica , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/terapia , Proliferação de Células/efeitos dos fármacos
2.
CNS Neurosci Ther ; 30(10): e70008, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39374157

RESUMO

BACKGROUND: Manganism, a central nervous system dysfunction correlated with neurological deficits such as Parkinsonism, is caused by the substantial collection of manganese chloride (MnCl2) in the brain. OBJECTIVES: To explore the neuroprotective effects of natural compounds, namely, micronized zeolite clinoptilolite (ZC) and punicalagin (PUN), either individually or in combination, against MnCl2-induced Parkinson's disease (PD). METHODS: Fifty male albino rats were divided into 5 groups (Gps). Gp I was used as the control group, and the remaining animals received MnCl2 (Gp II-Gp V). Rats in Gps III and IV were treated with ZC and PUN, respectively. Gp V received both ZC and PUN as previously reported for the solo-treated plants. RESULTS: ZC and/or PUN reversed the depletion of monoamines in the brain and decreased acetyl choline esterase activity, which primarily adjusted the animals' behavior and motor coordination. ZC and PUN restored the balance between glutamate/γ-amino butyric acid content and markedly improved the brain levels of brain-derived neurotrophic factor and nuclear factor erythroid 2-related factor 2/heme oxygenase-1 and decreased glycogen synthase kinase-3 beta activity. ZC and PUN also inhibited inflammatory and oxidative markers, including nuclear factor kappa-light-chain-enhancer of activated B cells, Toll-like receptor 4, nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 and caspase-1. Bcl-2-associated X-protein and B-cell leukemia/lymphoma 2 protein (Bcl-2) can significantly modify caspase-3 expression. ZC and/or PUN ameliorated PD in rats by decreasing the levels of endoplasmic reticulum (ER) stress markers (p-protein kinase-like ER kinase (PERK), glucose-regulated protein 78, and C/EBP homologous protein (CHOP)) and enhancing the levels of an autophagy marker (Beclin-1). DISCUSSION AND CONCLUSION: ZC and/or PUN mitigated the progression of PD through their potential neurotrophic, neurogenic, anti-inflammatory, antioxidant, and anti-apoptotic activities and by controlling ER stress through modulation of the PERK/CHOP/Bcl-2 pathway.


Assuntos
Cloretos , Taninos Hidrolisáveis , Fármacos Neuroprotetores , Zeolitas , Animais , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Zeolitas/farmacologia , Taninos Hidrolisáveis/farmacologia , Taninos Hidrolisáveis/uso terapêutico , Cloretos/toxicidade , Compostos de Manganês/farmacologia , Modelos Animais de Doenças , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo
3.
ACS Nano ; 18(41): 27853-27868, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39370780

RESUMO

Ultrahigh field magnetic resonance imaging (UHF-MRI) (≥7 T) can dramatically boost image resolution and signal-to-noise ratio, which have distinct advantages in multifunctional imaging. However, their research and application are currently limited by the absence of high-field contrast agents (CAs) and the low sensitivity and accuracy of T1/T2 single-modality CAs. Therefore, the development of T1-T2 dual-mode CAs that respond to UHF-MRI and nanoformulations with therapeutic sensitization can bring ideas for the integrated application of precise and synchronous tumor theranostics. Herein, we present a biomimetic mineralization strategy for synthesizing holmium/manganese oxide-bovine serum albumin-photosensitizer chlorin e6 nanohybrids. The hybrid nanoparticles exhibited better tumor accumulation, a suitable time imaging window, and excellent pH-response T1-T2 dual-mode UHF-MRI performance. The antitumor effect comes from the amelioration of the hypoxic tumor microenvironment to promote the synergistic effect of photodynamic therapy and radiotherapy, along with negligible acute toxicity. Undoubtedly, this work not only provides a different perspective for developing multifunctional nanotherapeutics but also promotes the potential clinical exploitation and translation of UHF CAs.


Assuntos
Biomineralização , Imageamento por Ressonância Magnética , Fármacos Fotossensibilizantes , Nanomedicina Teranóstica , Animais , Camundongos , Humanos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/síntese química , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Clorofilídeos , Fotoquimioterapia , Porfirinas/química , Porfirinas/farmacologia , Nanopartículas/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Óxidos/química , Soroalbumina Bovina/química , Meios de Contraste/química , Meios de Contraste/síntese química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Camundongos Endogâmicos BALB C
4.
J Nanobiotechnology ; 22(1): 664, 2024 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-39465387

RESUMO

Atherosclerosis (AS) poses a significant threat to human life and health. However, conventional antiatherogenic medications exhibit insufficient targeting precision and restricted therapeutic effectiveness. Moreover, during the progression of AS, macrophages undergo polarization toward the proinflammatory M1 phenotype and generate reactive oxygen species (ROS) to accelerate the occurrence of inflammatory storms, and ingest excess lipids to form foam cells by inhibiting cholesterol efflux. In our study, we developed a macrophage membrane-functionalized hollow mesoporous manganese dioxide nanomedicine (Col@HMnO2-MM). This nanomedicine has the ability to evade immune cell phagocytosis, enables prolonged circulation within the body, targets the inflammatory site of AS for effective drug release, and alleviates the inflammatory storm at the AS site by eliminating ROS. Furthermore, Col@HMnO2-MM has the ability to generate oxygen autonomously by breaking down surplus hydrogen peroxide generated at the inflammatory AS site, thereby reducing the hypoxic microenvironment of the plaque by downregulating hypoxia-inducible factor (HIF-1α), which in turn enhances cholesterol efflux to inhibit foam cell formation. In an APOE-/- mouse model, Col@HMnO2-MM significantly reduced inflammatory factor levels, lipid storage, and plaque formation without significant long-term toxicity. In summary, this synergistic treatment significantly improved the effectiveness of nanomedicine and may offer a novel strategy for precise AS therapy.


Assuntos
Aterosclerose , Colesterol , Macrófagos , Compostos de Manganês , Nanomedicina , Óxidos , Animais , Óxidos/química , Óxidos/farmacologia , Camundongos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Colesterol/metabolismo , Colesterol/química , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanomedicina/métodos , Espécies Reativas de Oxigênio/metabolismo , Inflamação/tratamento farmacológico , Células RAW 264.7 , Humanos , Camundongos Endogâmicos C57BL , Membrana Celular/metabolismo , Masculino , Células Espumosas/metabolismo , Células Espumosas/efeitos dos fármacos , Modelos Animais de Doenças
5.
ACS Appl Bio Mater ; 7(10): 6529-6541, 2024 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-39357930

RESUMO

Between 2015 and 2017, 90% of Chinese adults were reported to have periodontitis of varying degrees, highlighting the importance of novel, inexpensive, and affordable treatments for the public. The fact that more and more pathogens are becoming resistant to antibiotics further highlights this prevalence. This article addresses a novel micromotor capable of generating reactive oxygen species, as proven by a Fenton-like reaction. Such reactions allow the targeting of Gram-negative bacteria such as Escherichia coli, which are eliminated order of magnitude more effectively than by pure hydrogen peroxide, thereby addressing pathogens relevant in oral infections. The basis of the micromotors, which generate reactive oxygen species on site, reduces the likelihood of resistance developing in these types of bacteria. Catalytically reducing hydrogen peroxide in this process, these micromotors propel themselves forward. This proof of principle study paves the way for the utilization of micromotors in the field of skin disinfection utilizing hydrogen peroxide concentrations which were in previous works proven noncytotoxic.


Assuntos
Antibacterianos , Escherichia coli , Peróxido de Hidrogênio , Compostos de Manganês , Teste de Materiais , Testes de Sensibilidade Microbiana , Óxidos , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/química , Antibacterianos/farmacologia , Antibacterianos/química , Escherichia coli/efeitos dos fármacos , Óxidos/química , Óxidos/farmacologia , Catálise , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Tamanho da Partícula , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Espécies Reativas de Oxigênio/metabolismo
6.
ACS Appl Mater Interfaces ; 16(38): 50238-50250, 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39284745

RESUMO

Damaged skin is susceptible to invasion by harmful microorganisms, especially Staphylococcus aureus and Escherichia coli, which can delay healing. Epigallocatechin-3-gallate (EGCG) is a natural compound known for effectively promoting wound healing and its potent anti-inflammatory effects. However, its application is limited due to its susceptibility to oxidation and isomerization, which alter its structure. The use of zeolitic imidazolate framework-8 (ZIF-8) can effectively tackle these issues. This study introduces an oxygen (O2) and hydrogen peroxide (H2O2) self-supplying ZIF-8 nanoplatform designed to enhance the bioavailability of EGCG, combining photodynamic therapy (PDT) and chemodynamic therapy (CDT) to improve antibacterial properties and ultimately accelerate wound healing. For this purpose, EGCG and indocyanine green (ICG), a photosensitizer, were successively integrated into a ZIF-8, and coated with bovine serum albumin (BSA) to enhance biocompatibility. The outer layer of this construct was further modified with manganese dioxide (MnO2) to promote CDT and calcium peroxide (CaO2) to supply H2O2 and O2, resulting in the final nanoplatform EGCG-ICG@ZIF-8/BSA-MnO2/CaO2 (EIZBMC). In in vitro experiments under 808 nm laser, EIZBMC exhibited synergistic antibacterial effects through PDT and CDT. This combination effectively released reactive oxygen species (ROS), which mediated oxidative stress to inhibit the bacteria. Subsequently, in a murine model of wound infection, EIZBMC not only exerted antibacterial effects through PDT and CDT but also alleviated the inflammatory condition and promoted the regeneration of collagen fibers, which led to accelerated wound healing. Overall, this research presents a promising approach to enhancing the therapeutic efficacy of EGCG by leveraging the synergistic antibacterial effects of PDT and CDT. This multifunctional nanoplatform maximizes EGCG's anti-inflammatory properties, offering a potent solution for promoting infected wound healing.


Assuntos
Antibacterianos , Catequina , Escherichia coli , Verde de Indocianina , Fotoquimioterapia , Fármacos Fotossensibilizantes , Staphylococcus aureus , Cicatrização , Animais , Catequina/química , Catequina/análogos & derivados , Catequina/farmacologia , Cicatrização/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Camundongos , Verde de Indocianina/química , Verde de Indocianina/farmacologia , Escherichia coli/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Peróxido de Hidrogênio/farmacologia , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Óxidos/química , Óxidos/farmacologia , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Zeolitas/química , Zeolitas/farmacologia , Nanopartículas/química , Testes de Sensibilidade Microbiana , Imidazóis/química , Imidazóis/farmacologia , Peróxidos
7.
J Nanobiotechnology ; 22(1): 577, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300539

RESUMO

Treating bone defect concomitant with microbial infection poses a formidable clinical challenge. Addressing this dilemma necessitates the implementation of biomaterials exhibiting dual capabilities in anti-bacteria and bone regeneration. Of particular significance is the altered microenvironment observed in infected bones, characterized by acidity, inflammation, and an abundance of reactive oxygen species (ROS). These conditions, while challenging, present an opportunity for therapeutic intervention in the context of contaminated bone defects. In this study, we developed an oriented composite scaffold containing copper-coated manganese dioxide (MnO2) nanoparticles loaded with parathyroid hormone (PMPC/Gelatin). The characteristics of these scaffolds were meticulously evaluated and confirmed the high sensitivity to H+, responsive drug release and ROS elimination. In vitro antibacterial analysis underscored the remarkable ability of PMPC/Gelatin scaffolds to substantially suppressed bacterial proliferation and colony formation. Furthermore, this nontoxic material demonstrated efficacy in mitigating ROS levels, thereby fostering osteogenic differentiation of bone marrow mesenchymal stem cells and enhancing angiogenic ability. Subsequently, the infected models of bone defects in rat skulls were established to investigate the effects of composite scaffolds on anti-bacteria and bone formation in vivo. The PMPC/Gelatin treatment exhibited excellent antibacterial activity, coupled with enhanced vascularization and osteogenesis at the defect sites. These compelling findings affirm that the PMPC/Gelatin composite scaffold represents a promising avenue for anti-bacteria and bone regeneration.


Assuntos
Antibacterianos , Regeneração Óssea , Gelatina , Compostos de Manganês , Células-Tronco Mesenquimais , Osteogênese , Óxidos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Alicerces Teciduais , Animais , Osteogênese/efeitos dos fármacos , Alicerces Teciduais/química , Ratos , Regeneração Óssea/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Gelatina/química , Óxidos/química , Óxidos/farmacologia , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Masculino , Crânio/efeitos dos fármacos , Nanopartículas/química , Cobre/química , Cobre/farmacologia , Diferenciação Celular/efeitos dos fármacos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
8.
Biomed Pharmacother ; 179: 117402, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39243428

RESUMO

In recent years, the integration of radiotherapy and nanocatalytic medicine has gained widespread attention in the treatment of breast cancer. Herein, the glucose oxidase (GOx) and MnO2 nanoparticles co-modified multifunctional liposome of GOx-MnO2@Lip was constructed for enhanced radiotherapy. Introduction of GOx would not only elevate the glucose consumption to starve the cancer cells, but also increased the endogenous H2O2 level. Meanwhile, high intracellular GSH concentration facilitated the release of Mn2+ to amplify the cytotoxic ·OH through cascade catalytic reactions within the tumor microenvironment, resulting in a favorable tumor suppression rate of 74.45 %. Furthermore, the blood biochemical and blood routine demonstrated that GOx-MnO2@Lip had no obvious toxic side effects. Therefore, this work provided a potential vehicle for synergistic cancer starving therapy, chemodynamic therapy and radiotherapy for improving therapeutic efficacy of breast cancer.


Assuntos
Neoplasias da Mama , Glucose Oxidase , Lipossomos , Compostos de Manganês , Óxidos , Radiossensibilizantes , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Glucose Oxidase/metabolismo , Feminino , Óxidos/química , Óxidos/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Radiossensibilizantes/farmacologia , Animais , Humanos , Linhagem Celular Tumoral , Peróxido de Hidrogênio/metabolismo , Camundongos Endogâmicos BALB C , Catálise , Camundongos , Nanopartículas/química , Microambiente Tumoral/efeitos dos fármacos
9.
Nat Commun ; 15(1): 8058, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39277594

RESUMO

Pathogen-host competition for manganese and intricate immunostimulatory pathways severely attenuates the efficacy of antibacterial immunotherapy against biofilm infections associated with orthopaedic implants. Herein, we introduce a spatiotemporal sono-metalloimmunotherapy (SMIT) strategy aimed at efficient biofilm ablation by custom design of ingenious biomimetic metal-organic framework (PCN-224)-coated MnO2-hydrangea nanoparticles (MnPM) as a metalloantibiotic. Upon reaching the acidic H2O2-enriched biofilm microenvironment, MnPM can convert abundant H2O2 into oxygen, which is conducive to significantly enhancing the efficacy of ultrasound (US)-triggered sonodynamic therapy (SDT), thereby exposing bacteria-associated antigens (BAAs). Moreover, MnPM disrupts bacterial homeostasis, further killing more bacteria. Then, the Mn ions released from the degraded MnO2 can recharge immune cells to enhance the cGAS-STING signaling pathway sensing of BAAs, further boosting the immune response and suppressing biofilm growth via biofilm-specific T cell responses. Following US withdrawal, the sustained oxygenation promotes the survival and migration of fibroblasts, stimulates the expression of angiogenic growth factors and angiogenesis, and neutralizes excessive inflammation. Our findings highlight that MnPM may act as an immune costimulatory metalloantibiotic to regulate the cGAS-STING signaling pathway, presenting a promising alternative to antibiotics for orthopaedic biofilm infection treatment and pro-tissue repair.


Assuntos
Biofilmes , Compostos de Manganês , Óxidos , Oxigênio , Biofilmes/efeitos dos fármacos , Animais , Camundongos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Oxigênio/metabolismo , Óxidos/farmacologia , Óxidos/química , Antibacterianos/farmacologia , Peróxido de Hidrogênio/metabolismo , Imunoterapia/métodos , Humanos , Terapia por Ultrassom/métodos , Nanopartículas/química , Transdução de Sinais/efeitos dos fármacos , Antígenos de Bactérias/imunologia , Staphylococcus aureus/efeitos dos fármacos , Feminino
10.
ACS Nano ; 18(40): 27487-27502, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39329191

RESUMO

Reprogramming of cellular metabolism in tumors promoted the epithelial-mesenchymal transition (EMT) process and established immune-suppressive tumor microenvironments (iTME), leading to drug resistance and tumor progression. Therefore, remodeling the cellular metabolism of tumor cells was a promising strategy to overcome drug-resistant tumors. Herein, CD276 and MTHFD2 were identified as a specific marker and a therapeutic target, respectively, for targeting sunitinib-resistant clear cell renal cell carcinoma (ccRCC) and its cancer stem cell (CSC) population. The blockade of MTHFD2 was confirmed to overcome drug resistance via remodeling of folate-nucleotide metabolism. Moreover, the manganese dioxide nanoparticle was proven here by a high-throughput metabolome to be capable of remodeling γ-aminobutyric acid (GABA) metabolism in tumor cells to reconstruct the iTME. Based on these findings, engineered CD276-CD133 dual-targeting biomimetic nanovesicle EMφ-siMTHFD2-MnO2@Suni was designed to overcome drug resistance and terminate tumor progression of ccRCC. Using ccRCC-bearing immune-humanized NPG model mice, EMφ-siMTHFD2-MnO2@Suni was observed to remodel folate-nucleotide and GABA metabolism to deactivate the EMT process and reconstruct the iTME thereby overcoming the drug resistance. In the incomplete-tumor-resection recurrence model and metastasis model, EMφ-siMTHFD2-MnO2@Suni reduced recurrence and metastasis in vivo. This work thus provided an innovative approach that held great potential in the treatment of drug-resistant ccRCC by remodeling cellular metabolism.


Assuntos
Carcinoma de Células Renais , Resistencia a Medicamentos Antineoplásicos , Ácido Fólico , Neoplasias Renais , Compostos de Manganês , Sunitinibe , Ácido gama-Aminobutírico , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Animais , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Ácido Fólico/química , Ácido Fólico/metabolismo , Camundongos , Sunitinibe/farmacologia , Sunitinibe/química , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/química , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Nanopartículas/química , Nucleotídeos/química , Nucleotídeos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Materiais Biomiméticos/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Aminoidrolases , Metilenotetra-Hidrofolato Desidrogenase (NADP) , Óxidos , Enzimas Multifuncionais
11.
J Nanobiotechnology ; 22(1): 535, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227944

RESUMO

BACKGROUND: Manganese ions (Mn2+) combined with adjuvants capable of damaging and lysing tumor cells form an antitumor nano-modulator that enhances the immune efficacy of cancer therapy through the cascade activation of the cyclic GMP-AMP interferon gene synthase-stimulator (cGAS-STING) pathway, which underscores the importance of developing antitumor nano-modulators, which induce DNA damage and augment cGAS-STING activity, as a critical future research direction. METHODS AND RESULTS: We have successfully synthesized an antitumor nano-modulator, which exhibits good dispersibility and biosafety. This nano-modulator is engineered by loading manganese dioxide nanosheets (M-NS) with zebularine (Zeb), known for its immunogenicity-enhancing effects, and conducting targeted surface modification using hyaluronic acid (HA). After systemic circulation to the tumor site, Mn2+, Zeb, and reactive oxygen species (ROS) are catalytically released in the tumor microenvironment by H+ and H2O2. These components can directly or indirectly damage the DNA or mitochondria of tumor cells, thereby inducing programmed cell death. Furthermore, they promote the accumulation of double-stranded DNA (dsDNA) in the cytoplasm, enhancing the activation of the cGAS-STING signalling pathway and boosting the production of type I interferon and the secretion of pro-inflammatory cytokines. Additionally, Zeb@MH-NS enhances the maturation of dendritic cells, the infiltration of cytotoxic T lymphocytes, and the recruitment of natural killer cells at the tumor site. CONCLUSIONS: This HA-modified manganese-based hybrid nano-regulator can enhance antitumor therapy by boosting innate immune activity and may provide new directions for immunotherapy and clinical translation in cancer.


Assuntos
Imunidade Inata , Compostos de Manganês , Proteínas de Membrana , Nucleotidiltransferases , Óxidos , Transdução de Sinais , Microambiente Tumoral , Nucleotidiltransferases/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Animais , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Proteínas de Membrana/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , Óxidos/química , Óxidos/farmacologia , Manganês/química , Manganês/farmacologia , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/química , Linhagem Celular Tumoral , Espécies Reativas de Oxigênio/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Feminino , Camundongos Endogâmicos C57BL
12.
J Photochem Photobiol B ; 259: 113002, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39141980

RESUMO

The hypoxic environment within a solid tumor is a limitation to the effectiveness of photodynamic therapy. Here, we demonstrate the use of oxygen generating nanozymes (CeO2, Fe3O4, and MnO2) to improve the photodynamic effect. The optimized combination of process parameters for irradiation was obtained using the Box Behnken experimental design. Indocyanine green, IR 820, and their different combinations with oxygen generators were studied for their effect on oral carcinoma. Dynamic light scattering technique showed the average particle size of CeO2, MnO2, and Fe3O4 to be 211 ± 16, and 157 ± 28, 143 ± 19 nm with PDI of 0.23, 0.28 and 0.20 and a zeta potential of -2.6 ± 0.45, -2.4 ± 0.60 and  -6.1 ± 0.23 mV, respectively. The formation of metal oxides was confirmed using UV-visible, FTIR, and X-ray photon spectroscopies. The amount of dissolved oxygen produced by CeO2, MnO2, and Fe3O4 in the presence of H2O2 within 2 min was 1.7 ± 0.15, 1.7 ± 0.16, and 1.4 ± 0.12 mg/l, respectively. Growth inhibition studies in the FaDu oral carcinoma spheroid model showed a significant (P < 0.05) increase in growth reduction from 81 ± 2.9 and 88 ± 2.1% to 97 ± 1.2 and 99 ± 1.0% for ICG and IR 820, respectively, after irradiation (808 nm laser, 1 W/cm2, 5 min) in the presence of CeO2 (25 µg/ml). In conclusion, oxygen-generating nanozymes can improve the photodynamic effect of ICG and IR 820.


Assuntos
Cério , Verde de Indocianina , Compostos de Manganês , Neoplasias Bucais , Óxidos , Oxigênio , Verde de Indocianina/química , Verde de Indocianina/farmacologia , Humanos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Oxigênio/química , Oxigênio/metabolismo , Óxidos/química , Óxidos/farmacologia , Cério/química , Cério/farmacologia , Linhagem Celular Tumoral , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Peróxido de Hidrogênio/química , Tamanho da Partícula , Sobrevivência Celular/efeitos dos fármacos
13.
Mil Med ; 189(Suppl 3): 598-607, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39160887

RESUMO

INTRODUCTION: Military members and first responders may, at moment's notice, be asked to assist in incidents that may result in radiation exposure such as Operation Tomadachi in which the U.S. Navy provided significant relief for the Fukushima Daiichi Nuclear Reactor accident in Japan after an earthquake and tsunami in 2011. We are also currently facing potential threats from nuclear power plants in the Ukraine should a power disruption to a nuclear plant interfere with cooling or other safety measures. Exposure to high doses of radiation results in acute radiation syndrome (ARS) characterized by symptoms arising from hematopoietic, gastrointestinal, and neurovascular injuries. Although there are mitigators FDA approved to treat ARS, there are currently no FDA-approved prophylactic medical interventions to help protect persons who may need to respond to radiation emergencies. There is strong evidence that manganese (Mn) has radiation protective efficacy as a promising prophylactic countermeasure. MATERIALS AND METHODS: All animal procedures were approved by the Institutional Animal Care and Use Committee. Male and female B6D2F1J mice, 10 to 11 weeks old, were used for neurotoxicity studies and temporal effects of Mn. Four groups were evaluated: (1) vehicle injection, (2) dose of 4.5 mg/kg for 3 days, (3) dose of 13.5 mg/kg, and (4) sham. Irradiated mice were exposed to 9.5 Gy whole body Co60 γ-radiation. MRI was performed with a high dose of manganese chloride (MnCl2) (150 mg/kg) to assess the distribution of the MnCl2. RESULTS: The mice have promising survival curves (highest survival-13.5 mg/kg dose over 3 days of MnCl2 at 80% [87% female, 73% male] P = 0.0004). The complete blood count (CBC) results demonstrated a typical hematopoietic response in all of the irradiated groups, followed by mildly accelerated recovery by day 28 in the treated groups. No difference between groups was measured by Rota Rod, DigiGait, and Y-maze. Histologic evaluation of the bone marrow sections in the group given 13.5 mg/kg dose over 3 days had the best return to cellularity at 80%. MRI showed a systemic distribution of MnCl2. DISCUSSION: The preliminary data suggest that a dose of 13.5 mg/kg of MnCl2 given over 3 days prior to exposure of radiation may have a protective benefit while not exhibiting the neurobehavioral problems. A countermeasure that can prophylactically protect emergency personnel entering an area contaminated with high levels of radiation is needed, especially in light that nuclear accidents are a continued global threat. There is a need for a protective agent with easy long-term storage, easy to transport, easy to administer, and low cost. Histologic evaluation supports the promising effect of MnCl2 in protecting tissue, especially the bone marrow using the dose given over 3 days (4.5 mg/kg per day) of MnCl2. CONCLUSIONS: Initial experiments show that MnCl2 is a promising safe and effective prophylactic countermeasure against ARS. MRI data support the systemic distribution of MnCl2 which is needed in order to protect multiple tissues in the body. The pathology data in bone marrow and the brain support faster recovery from radiation exposure in the treated animals and decreased organ damage.


Assuntos
Síndrome Aguda da Radiação , Cloretos , Compostos de Manganês , Protetores contra Radiação , Síndrome Aguda da Radiação/prevenção & controle , Síndrome Aguda da Radiação/tratamento farmacológico , Animais , Camundongos , Feminino , Masculino , Compostos de Manganês/farmacologia , Protetores contra Radiação/farmacologia , Protetores contra Radiação/uso terapêutico
14.
Acta Biomater ; 187: 366-380, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39209133

RESUMO

Ferroptosis is greatly restricted by low reactive oxygen species (ROS) generation efficiency, and the inherent self-protection mechanism originating in heat shock proteins (HSPs) seriously impedes the efficiency of photothermal therapy (PTT). Herein, we designed an intelligent strategy utilizing cascade catalytic nanoassemblies (Au@COF@MnO2) with triple-enzyme activity for amplifying ferroptosis therapy and improving the efficiency of PTT in tumor. Gold nanozyme was encapsulated within a hollow manganese dioxide (MnO2) shell with the help of covalent organic frameworks (COFs). The nanoassemblies possess the ability of photothermal conversion. Mechanism studies suggested that glutathione (GSH) depletion by Au@COF@MnO2 leads to the inactivation of glutathione peroxidase 4 (GPX4). This effect synergized with Mn2+-mediated reactive oxygen species (ROS) generation to enhance the accumulation of lipid peroxide (LPO), thereby inducing high-efficiency ferroptosis. Notably, gold nanozyme facilitated the conversion of glucose into gluconic acid and hydrogen peroxide (H2O2). This process augmented the endogenous H2O2 levels necessary for Fenton chemistry, which could effectively promote the generation of ROS. Simultaneously, glucose depletion downregulated the expression of HSPs induced by hyperthermia, consequently reducing cellular heat resistance for enhancing PTT. Therefore, the cascade catalytic nanoassembly not only exhibits high tumor inhibition and admirable biosafety, but also possesses trimodal imaging performance for imaging-guided tumor therapy in vivo, holding great potential for clinical application. STATEMENT OF SIGNIFICANCE: This study engineered multi-responsive cascade catalytic nanoassembly (Au@COF@MnO2) with triple enzymatic functions for amplifying ferroptosis therapy and improving the efficiency of PTT in tumor. The nanoassembly exhibited multi-responsive release and great photothermal conversion performance. Glucose consumption-evoked starvation downregulated the hyperthermia-induced expression of HSPs in tumor cells, thereby improving the efficacy of PTT. Mechanism studies suggested that GSH depletion by Au@COF@MnO2 lead to the inactivation of GPX4, which synergized with Mn2+-mediated ROS generation to bolster the accumulation of LPO, thereby inducing high-efficiency ferroptosis. Moreover, the nanoassembly demonstrated trimodal (PT, PA, and MR) imaging in vivo, enabling the visualization of the tumor treatment with nanoassembly. Such nanoassembly exhibited high tumor inhibition and admirable biosafety in tumor therapy in vivo, holding a great potential for clinical application.


Assuntos
Ferroptose , Ouro , Compostos de Manganês , Óxidos , Terapia Fototérmica , Ferroptose/efeitos dos fármacos , Animais , Humanos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Ouro/química , Ouro/farmacologia , Camundongos , Óxidos/química , Óxidos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Catálise , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Camundongos Nus , Feminino , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio
15.
J Colloid Interface Sci ; 676: 378-395, 2024 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-39032420

RESUMO

Glioma is a prevalent brain malignancy associated with poor prognosis. Although chemotherapy serves as the primary treatment for brain tumors, its effectiveness is hindered by the limited ability of drugs to traverse the blood-brain barrier (BBB) and the development of drug resistance linked to tumor hypoxia. Herein, we report the creation of hybrid camouflaged multifunctional nanovesicles comprising membranes of tumor C6 cells (mT) and bacterial outer membrane vesicles (OMVs) and co-loaded with manganese dioxide nanoparticles (MnO2 NPs) and doxorubicin (DOX) to synergistically enhance the chemotherapy/chemodynamic therapy (CDT) of glioma. Owing to OMV-mediated BBB penetration and mT-inherited tumor-homing properties, MnO2-DOX@mT/OMVs can penetrate the BBB and enhance the tumor cell-specific uptake of DOX via "proton sponge effect"-mediated lysosomal escape. This enhances the apoptotic effect induced by DOX and minimizing DOX-associated cardiotoxicity by facilitating the accumulation of DOX at the tumor site. Furthermore, the MnO2 NPs in MnO2-DOX@mT/OMVs can generate potent CDT by accelerating the Fenton-like reaction with DOX-generated H2O2 and achieving glutathione (GSH)-depletion-induced glutathione peroxidase 4 (GPX4) inactivation. These results showed that MnO2-DOX@mT/OMVs, designed for brain tumor targeting, significantly inhibited tumor growth and exhibited favorable biological safety. This innovative approach offers the augmentation of anticancer treatment efficacy via a potential combination of chemotherapy and CDT.


Assuntos
Neoplasias Encefálicas , Doxorrubicina , Glioma , Compostos de Manganês , Nanopartículas , Óxidos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Óxidos/química , Óxidos/farmacologia , Glioma/tratamento farmacológico , Glioma/patologia , Glioma/metabolismo , Doxorrubicina/farmacologia , Doxorrubicina/química , Animais , Nanopartículas/química , Camundongos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/química , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Membrana Celular/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/química , Linhagem Celular Tumoral , Humanos , Tamanho da Partícula , Ensaios de Seleção de Medicamentos Antitumorais , Sobrevivência Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Propriedades de Superfície , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Ratos
16.
J Colloid Interface Sci ; 676: 1088-1097, 2024 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-39079272

RESUMO

Bimetallic nanozymes exhibited multi-enzyme activities, but glutathione (GSH) overexpression and weak catalytic capability restricted their catalytic therapeutic performance. Thus, this study developed a smart nanozyme (AuPt@MnO2) with a core-shell structure by coating manganese dioxide (MnO2) on the gold-platinum (AuPt) nanozyme (AuPt@MnO2) surface to enhance catalytic therapy. In this nanozyme, AuPt possessed triple-enzyme activities, i.e., catalase, peroxidase, and glucose oxidase, which greatly improved oxygen, hydroxyl radicals (·OH), and hydrogen peroxide generation, due to cyclic reactions. Moreover, GSH consumption degraded the MnO2 shell, which then enhanced ·OH generation of Mn2+. More importantly, the near-infrared-II (NIR-II) photothermal performance of AuPt@MnO2 with a high conversion efficiency of 38.7 % further promoted multi-enzyme activities and enhanced catalytic therapy. Moreover, combining NIR-II photothermal therapy and enhancing catalytic therapy decreased the cell viability to 10.8 %, and thereby, the tumors were cleared. Thus, the AuPt@MnO2 smart nanoplatform developed in this study exhibited NIR-II photothermal-promoted multi-enzyme activities and excellent antitumor efficacy, which will be promising for enhancing catalytic therapy.


Assuntos
Catalase , Sobrevivência Celular , Ouro , Raios Infravermelhos , Compostos de Manganês , Óxidos , Platina , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Platina/química , Platina/farmacologia , Ouro/química , Ouro/farmacologia , Catálise , Óxidos/química , Óxidos/farmacologia , Humanos , Animais , Sobrevivência Celular/efeitos dos fármacos , Catalase/química , Catalase/metabolismo , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/química , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Terapia Fototérmica , Propriedades de Superfície , Peroxidase/metabolismo , Peroxidase/química , Tamanho da Partícula
17.
J Am Chem Soc ; 146(31): 21568-21582, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39051165

RESUMO

The heterogeneity of hepatocellular carcinoma (HCC) can prevent effective treatment, emphasizing the need for more effective therapies. Herein, we employed arsenene nanosheets coated with manganese dioxide and polyethylene glycol (AMPNs) for the degradation of Pin1, which is universally overexpressed in HCC. By employing an "AND gate", AMPNs exhibited responsiveness toward excessive glutathione and hydrogen peroxide within the tumor microenvironment, thereby selectively releasing AsxOy to mitigate potential side effects of As2O3. Notably, AMPNs induced the suppressing Pin1 expression while simultaneously upregulation PD-L1, thereby eliciting a robust antitumor immune response and enhancing the efficacy of anti-PD-1/anti-PD-L1 therapy. The combination of AMPNs and anti-PD-1 synergistically enhanced tumor suppression and effectively induced long-lasting immune memory. This approach did not reveal As2O3-associated toxicity, indicating that arsenene-based nanotherapeutic could be employed to amplify the response rate of anti-PD-1/anti-PD-L1 therapy to improve the clinical outcomes of HCC patients and potentially other solid tumors (e.g., breast cancer) that are refractory to anti-PD-1/anti-PD-L1 therapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Compostos de Manganês , Peptidilprolil Isomerase de Interação com NIMA , Óxidos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Óxidos/química , Óxidos/farmacologia , Humanos , Peptidilprolil Isomerase de Interação com NIMA/antagonistas & inibidores , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Nanoestruturas/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Arsenicais/química , Arsenicais/farmacologia , Arsenicais/uso terapêutico , Camundongos , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Linhagem Celular Tumoral , Polietilenoglicóis/química
18.
Molecules ; 29(13)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38998912

RESUMO

Managing chronic non-healing wounds presents a significant clinical challenge due to their frequent bacterial infections. Mesoporous silica-based materials possess robust wound-healing capabilities attributed to their renowned antimicrobial properties. The current study details the advancement of mesoporous silicon-loaded MnO and CaO molecules (HMn-Ca) against bacterial infections and chronic non-healing wounds. HMn-Ca was synthesized by reducing manganese chloride and calcium chloride by urotropine solution with mesoporous silicon as the template, thereby transforming the manganese and calcium ions on the framework of mesoporous silicon. The developed HMn-Ca was investigated using scanning electron microscopy (SEM), transmission electron microscope (TEM), ultraviolet-visible (UV-visible), and visible spectrophotometry, followed by the determination of Zeta potential. The production of reactive oxygen species (ROS) was determined by using the 3,3,5,5-tetramethylbenzidine (TMB) oxidation reaction. The wound healing effectiveness of the synthesized HMn-Ca is evaluated in a bacterial-infected mouse model. The loading of MnO and CaO inside mesoporous silicon enhanced the generation of ROS and the capacity of bacterial capture, subsequently decomposing the bacterial membrane, leading to the puncturing of the bacterial membrane, followed by cellular demise. As a result, treatment with HMn-Ca could improve the healing of the bacterial-infected wound, illustrating a straightforward yet potent method for engineering nanozymes tailored for antibacterial therapy.


Assuntos
Compostos de Manganês , Nanopartículas , Óxidos , Espécies Reativas de Oxigênio , Cicatrização , Cicatrização/efeitos dos fármacos , Animais , Camundongos , Nanopartículas/química , Óxidos/química , Óxidos/farmacologia , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Porosidade , Espécies Reativas de Oxigênio/metabolismo , Anti-Infecciosos/farmacologia , Anti-Infecciosos/química , Compostos de Cálcio/química , Compostos de Cálcio/farmacologia , Oxirredução , Antibacterianos/farmacologia , Antibacterianos/química , Manganês/química , Manganês/farmacologia , Testes de Sensibilidade Microbiana
19.
Sci Rep ; 14(1): 15436, 2024 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965280

RESUMO

Alzheimer disease (AD) is the cause of dementia and accounts for 60-80% cases. Tumor Necrosis Factor-alpha (TNF-α) is a multifunctional cytokine that provides resistance to infections, inflammation, and cancer. It developed as a prospective therapeutic target against multiple autoimmune and inflammatory disorders. Cholinergic insufficiency is linked to Alzheimer's disease, and several cholinesterase inhibitors have been created to treat it, including naturally produced inhibitors, synthetic analogs, and hybrids. In the current study, we tried to prepared compounds may also support the discovery and development of novel therapeutic and preventative drugs for Alzheimer's using manganese tetroxide nanoparticles (Mn3O4-NPs) as a catalyst to generate compounds with excellent reaction conditions. The Biginelli synthesis yields 4-(4-cyanophenyl)-6-oxo-2-thioxohexahydropyrimidine-5-carbonitrile when the 4-cyanobenzaldehyde, ethyl cyanoacetate, and thiourea were coupled with Mn3O4-NPs to produce compound 1. This multi-component method is non-toxic, safe, and environmentally friendly. The new approach reduced the amount of chemicals used and preserved time. Compound 1 underwent reactions with methyl iodide, acrylonitrile, chloroacetone, ethyl chloroacetate, and chloroacetic acid/benzaldehyde, each of the synthetized compounds was docked with TNF-α converting enzyme. These compounds may also support the discovery and development of novel therapeutic and preventative drugs for Alzheimer's disease. The majority of the produced compounds demonstrated pharmacokinetic features, making them potentially attractive therapeutic candidates for Alzheimer's disease treatment.


Assuntos
Doença de Alzheimer , Compostos de Manganês , Simulação de Acoplamento Molecular , Nanopartículas , Óxidos , Pirimidinas , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Pirimidinas/química , Pirimidinas/farmacologia , Pirimidinas/farmacocinética , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Animais , Nanopartículas/química , Óxidos/química , Óxidos/farmacologia , Humanos , Ratos , Masculino
20.
Bioorg Chem ; 150: 107593, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38971093

RESUMO

Nitric oxide (NO) and reactive oxygen species (ROS) embody excellent potential in cancer therapy. However, as a small molecule, their targeted delivery and precise, controllable release are urgently needed to achieve accurate cancer therapy. In this paper, a novel US-responsive bifunctional molecule (SD) and hyaluronic acid-modified MnO2 nanocarrier was developed, and a US-responsive NO and ROS controlled released nanoplatform was constructed. US can trigger SD to release ROS and NO simultaneously at the tumor site. Thus, SD served as acoustic sensitizer for sonodynamic therapy and NO donor for gas therapy. In the tumor microenvironment, the MnO2 nanocarrier can effectively deplete the highly expressed GSH, and the released Mn2+ can make H2O2 to produce .OH by Fenton-like reaction, which exhibited a strong chemodynamic effect. The high concentration of ROS and NO in cancer cell can induce cancer cell apoptosis ultimately. In addition, toxic ONOO-, which was generated by the reaction of NO and ROS, can effectively cause mitochondrial dysfunction, which induced the apoptosis of tumor cells. The 131I was labeled on the nanoplatform, which exhibited internal radiation therapy for tumor therapy. In -vitro and -vivo experiments showed that the nanoplatform has enhanced biocompatibility, and efficient anti-tumor potential, and it achieves synergistic sonodynamic/NO/chemodynamic/radionuclide therapy for cancer.


Assuntos
Radioisótopos do Iodo , Compostos de Manganês , Óxido Nítrico , Óxidos , Espécies Reativas de Oxigênio , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Humanos , Animais , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Óxidos/química , Óxidos/farmacologia , Radioisótopos do Iodo/química , Apoptose/efeitos dos fármacos , Nanopartículas/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Camundongos , Ensaios de Seleção de Medicamentos Antitumorais , Proliferação de Células/efeitos dos fármacos , Estrutura Molecular , Camundongos Endogâmicos BALB C , Terapia por Ultrassom , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ondas Ultrassônicas , Linhagem Celular Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...