Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Drug Deliv Transl Res ; 14(7): 1954-1968, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38191781

RESUMO

To improve treatment compliance and reach sustained and controlled drug release in the colon, we developed a hollow mesoporous silica nano-suppository that responded to both pH and redox stimuli. Firstly, we prepared hollow mesoporous silica nanoparticles containing disulfide bonds (HMSN-SS) and loaded them with 5-ASA. Secondly, we modified the surface of HMSN-SS with polydopamine (PDA) and chitosan (CS) and molded the suppository, which we named 5-ASA@HMSN-SS-PDA-CS (5-ASA@HSPC). By administering 5-ASA@HSPC rectally, it acted directly on the affected area. CS helped the nanoparticles adhere to the colon's surface, while PDA dissociates from HMSN-SS due to protonation in the acidic environment of the ulcerative colon. The disulfide bonds were destroyed by the reducing environment of the colon, leading to a stable and slow release of encapsulated 5-ASA from the pores of HMSN. Finally, in vitro release experiments and in vivo pharmacokinetic and pharmacodynamic experiments had demonstrated that 5-ASA@HSPC exhibited a slow and steady action at the colonic site, with an excellent safety profile. This novel approach showed great potential in the treatment of ulcerative colitis.


Assuntos
Quitosana , Colite Ulcerativa , Liberação Controlada de Fármacos , Indóis , Mesalamina , Nanopartículas , Oxirredução , Polímeros , Dióxido de Silício , Colite Ulcerativa/tratamento farmacológico , Concentração de Íons de Hidrogênio , Quitosana/química , Quitosana/administração & dosagem , Animais , Nanopartículas/química , Nanopartículas/administração & dosagem , Mesalamina/química , Mesalamina/administração & dosagem , Mesalamina/farmacocinética , Dióxido de Silício/química , Dióxido de Silício/administração & dosagem , Polímeros/química , Polímeros/administração & dosagem , Indóis/administração & dosagem , Indóis/química , Indóis/farmacocinética , Supositórios/química , Masculino , Preparações de Ação Retardada/química , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Colo/efeitos dos fármacos , Colo/metabolismo , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacocinética , Porosidade
2.
Food Chem Toxicol ; 182: 114198, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37995826

RESUMO

N,N-dimethylformamide (DMF), a widely consumed industrial solvent with persistent characteristics, can induce occupational liver damage and pose threats to the general population due to the enormous DMF-containing industrial efflux and emission from indoor facilities. This study was performed to explore the roles of allyl methyl disulfide (AMDS) in liver damage induced by DMF and the underlying mechanisms. AMDS was found to effectively suppress the elevation in the liver weight/body weight ratio and serum aminotransferase activities, and reduce the mortality of mice induced by DMF. In addition, AMDS abrogated DMF-elicited increases in malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) levels and decreases in glutathione (GSH) levels in mouse livers. The increase in macrophage number, mRNA expression of M1 macrophage biomarkers, and protein expression of key components in the NF-κB pathway and NLRP3 inflammasome induced by DMF exposure were all suppressed by AMDS in mouse livers. Furthermore, AMDS inhibited DMF-induced cell damage and NF-κB activation in cocultured AML12 hepatocytes and J774A.1 macrophages. However, AMDS per se did not significantly affect the protein level and activity of CYP2E1. Collectively, these results demonstrate that AMDS effectively ameliorates DMF-induced acute liver damage possibly by suppressing oxidative stress and inactivating the NF-κB pathway and NLRP3 inflammasome.


Assuntos
Inflamassomos , Hepatopatias , Humanos , Camundongos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , NF-kappa B/metabolismo , Dimetilformamida/toxicidade , Dimetilformamida/metabolismo , Hepatopatias/metabolismo , Estresse Oxidativo , Fígado , Glutationa/metabolismo
3.
J Mater Chem B ; 11(43): 10395-10403, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37876312

RESUMO

We have developed a targeted nano-drug delivery system that effectively harnesses the anti-tumor properties of trifluoperazine (TFP), while concurrently mitigating its side effects on the central nervous system. The manufacturing process entailed the preparation of mesoporous silica nanoparticles (MSN-NH2), followed by the loading of trifluoperazine into the pores of MSN-NH2 and then surface modification with polyethylene glycol (PEG) and anisamide (AA), resulting in the formation of TFP@MSN@PEG-AA (abbreviated as TMPA) nanoparticles. In vitro and in vivo anti-tumor activity and hemolysis experiments showed that TMPA had an excellent safety profile and a good anti-tumor effect. Importantly, the drug content of the TMPA nanoparticle group was found to be significantly lower than that of the TFP group in the mouse brain tissue as determined by High Performance Liquid Chromatography (HPLC) detection. Therefore, the developed drug delivery system achieved the goal of maintaining TFP's anti-tumor action while avoiding its negative effects on the central nervous system.


Assuntos
Nanopartículas , Neoplasias , Camundongos , Animais , Dióxido de Silício/química , Trifluoperazina , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química
4.
Ecotoxicol Environ Saf ; 238: 113609, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35551047

RESUMO

N,N-dimethylformamide (DMF) is a non-negligible volatile hazardous material in indoor and outdoor environments. Although the hepatotoxicity of DMF has been well recognized, the underlying mechanisms remain unclear and prophylactic medicine is still lacking. Herein, we established a DMF-induced acute liver injury mouse model and investigated the underlying mechanisms focusing on oxidative stress and the nucleotide-binding domain and leucine-rich repeat receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome. DMF was found to induce oxidative stress, evidenced by the elevation of hepatic malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) adducts levels, and the decline of reduced glutathione (GSH) levels. However, neither N-acetyl cysteine (NAC) nor sulforaphane (SF) ameliorated the hepatoxicity induced by DMF in mice. Interestingly, DMF exposure led to focal necrosis of hepatocytes and NLRP3 inflammasome activation before the onset of obvious liver damage. In addition, DMF exposure induced infiltration and proinflammatory/M1 polarization of macrophages in mice livers. Furthermore, the inactivation of hepatic macrophages by GdCl3 significantly suppressed DMF-induced elevation of serum aminotransferase activities, neutrophile infiltration, and activation of NLRP3 inflammasome in mice liver. Collectively, these results suggest that DMF-induced acute hepatotoxicity may be attributed to the activation of NLRP3 inflammasome in liver macrophages, but not oxidative stress.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Hepatopatias , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Dimetilformamida , Inflamassomos , Fígado , Macrófagos , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR
5.
Biomaterials ; 285: 121517, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35504179

RESUMO

Systemic lupus erythematosus (SLE) is a potentially life-threatening autoimmune disease that is characterized by alterations in the balance between effector and regulatory CD4+ T cells. We observed the upregulation of the immune checkpoints (ICs) PD-1 and TIGIT in pathogenic CD4+ T cells during disease progression, and downregulation of their ligands PD-L1 and CD155. Inspired by biomimetic nanotechnology, we fabricated dexamethasone (DXM)-loaded IFN-γ-treated MHC class I deficient cancer membrane-coated nanoparticles (IM-MNPs/DXM) to safely harness the immunosuppressive power of tumor cells for the treatment of SLE. The IM-MNPs inherited the membrane functions, which allowed these particles to evade immune clearance and accumulate in inflammatory organs. The IM-MNPs specifically targeted SLE CD4+ T cells and agonist PD-1/TIGIT signaling to inhibit effector T cell function while enhancing the immunosuppressive function of regulatory T cells (Tregs). The sustained release of DXM inhibited the production of proinflammatory cytokines in the inflammatory microenvironment to further promote Treg-mediated immune homeostasis. The IM-MNPs/DXM showed significant therapeutic efficacy in ameliorating lupus nephritis (LN) and decreasing side effects in vivo. Therefore, the particle represents a promising platform to improve current SLE treatment efficacy while minimizing systemic side effects of DXM and ICs agonist therapy.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Nanopartículas , Dexametasona/uso terapêutico , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Receptor de Morte Celular Programada 1 , Receptores Imunológicos , Linfócitos T Reguladores
6.
Cell Mol Life Sci ; 79(4): 201, 2022 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-35325321

RESUMO

Alcoholic liver disease (ALD) is a global public health challenge due to the high incidence and lack of effective therapeutics. Evidence from animal studies and ALD patients has demonstrated that iron overload is a hallmark of ALD. Ethanol exposure can promote iron absorption by downregulating the hepcidin expression, which is probably mediated by inducing oxidative stress and promoting erythropoietin (EPO) production. In addition, ethanol may enhance iron uptake in hepatocytes by upregulating the expression of transferrin receptor (TfR). Iron overload in the liver can aggravate ethanol-elicited liver damage by potentiating oxidative stress via Fenton reaction, promoting activation of Kupffer cells (KCs) and hepatic stellate cells (HSCs), and inducing a recently discovered programmed iron-dependent cell death, ferroptosis. This article reviews the current knowledge of iron metabolism, regulators of iron homeostasis, the mechanism of ethanol-induced iron overload, detrimental effects of iron overload in the liver, and potential therapeutic targets.


Assuntos
Sobrecarga de Ferro , Hepatopatias Alcoólicas , Animais , Hepatócitos/metabolismo , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/terapia , Hepatopatias Alcoólicas/metabolismo
7.
J Nanobiotechnology ; 20(1): 50, 2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35078498

RESUMO

BACKGROUND: Although cisplatin-based chemotherapy has been used as the first-line treatment for ovarian cancer (OC), tumor cells develop resistance to cisplatin during treatment, causing poor prognosis in OC patients. Studies have demonstrated that overactivation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway is involved in tumor chemoresistance and that overexpression of microRNA-497 (miR497) may overcome OC chemotherapy resistance by inhibiting the mTOR pathway. However, the low transcriptional efficiency and unstable chemical properties of miR497 limit its clinical application. Additionally, triptolide (TP) was confirmed to possess a superior killing effect on cisplatin-resistant cell lines, partially through inhibiting the mTOR pathway. Even so, the clinical applications of TP are restricted by serious systemic toxicity and weak water solubility. RESULTS: Herein, whether the combined application of miR497 and TP could further overcome OC chemoresistance by synergically suppressing the mTOR signaling pathway was investigated. Bioinspired hybrid nanoparticles formed by the fusion of CD47-expressing tumor exosomes and cRGD-modified liposomes (miR497/TP-HENPs) were prepared to codeliver miR497 and TP. In vitro results indicated that the nanoparticles were efficiently taken up by tumor cells, thus significantly enhancing tumor cell apoptosis. Similarly, the hybrid nanoparticles were effectively enriched in the tumor areas and exerted significant anticancer activity without any negative effects in vivo. Mechanistically, they promoted dephosphorylation of the overactivated PI3K/AKT/mTOR signaling pathway, boosted reactive oxygen species (ROS) generation and upregulated the polarization of macrophages from M2 to M1 macrophages. CONCLUSION: Overall, our findings may provide a translational strategy to overcome cisplatin-resistant OC and offer a potential solution for the treatment of other cisplatin-resistant tumors.


Assuntos
Exossomos , MicroRNAs , Nanopartículas , Neoplasias Ovarianas , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Diterpenos , Resistencia a Medicamentos Antineoplásicos , Compostos de Epóxi , Exossomos/metabolismo , Humanos , Lipossomos/farmacologia , MicroRNAs/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Fenantrenos , Fosfatidilinositol 3-Quinases/metabolismo
8.
Onco Targets Ther ; 15: 87-101, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35087279

RESUMO

PURPOSE: Stichoposide C (STC) is a triterpene glycoside isolated from Thelenota ananas, which is previously demonstrated to wide spectrum of anticancer effects against various tumor cells. However, the antitumor effects and underlying molecular mechanisms in ovarian cancer (OC) cells are not fully understood. Here, we examined if and through which mechanisms STC exerts anticancer effects on OC. METHODS: CCK-8 and colony formation assays were used to detect cell viability and proliferation. Flow cytometry was used to detect apoptosis and cell cycle arrest. Protein expression and phosphorylation were measured by Western blotting analysis. Confocal fluorescence microscopy was used to observe the autophagy flux. Autophagosome formation was observed via transmission electron microscopy. Antitumor effect of STC was investigated in patient-derived organoids (PDOs) and A2780 subcutaneous xenograft tumors. RESULTS: STC was found that not only exerted antiproliferation activity and apoptosis but also induced autophagy. Mechanistically, STC induced autophagy via inhibited the AKT/mTOR signaling pathway in ovarian cancer cells. In addition, STC and an autophagy inhibitor 3-methyladenine (3-MA) combination treatment showed significant synergetic effects on inhibiting proliferation and promoting apoptosis in vitro. Consistent with cell experiments, STC also inhibited the growth of two OC PDOs. Finally, STC markedly reduced the growth of A2780 subcutaneous xenograft tumors without organ toxicity and activated autophagy in vivo. CONCLUSION: Stichoposide C exerts in vitro and in vivo anticancer effects on ovarian cancer by inducing autophagy via inhibiting AKT/mTOR pathway. The findings warrant further prove for STC as a potential therapeutic agent for ovarian cancer.

9.
J Biomed Nanotechnol ; 17(3): 426-438, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33875077

RESUMO

Ovarian cancer has been the most lethal gynaecological malignancy worldwide. Additionally, triptolide is an active substance that has been extracted from the Chinese herbal medicine T. wilfordii Hook F. , which possesses anti-tumor, immunomodulatory and anti-inflammatory properties. In recent years, TP has attracted increasing attention because of its broad-spectrum efficient anti-tumor activity. Nevertheless, its clinical utility is limited due to its severe side effects. In this study, we constructed an exosome-encapsulated TP targeted drug delivery systems, studying its anti-tumor effects and mechanisms in vivo and in vitro . We observed that compared with free TP, TP-Exos significantly enhanced anti-ovarian cancer effects and reduce toxicity to important organs. We further demonstrated that TP-Exos induced apoptosis of ovarian cancer cells, regulated tumor immunity by activating the mitochondrial apoptosis pathway and selectively inhibited M2 tumor-associated macrophages and their tumor-promoting mediators in the tumor microenvironment. In summary, TP-Exos are a promising treatment for ovarian cancer.


Assuntos
Diterpenos , Exossomos , Neoplasias Ovarianas , Fenantrenos , Apoptose , Compostos de Epóxi , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Microambiente Tumoral
10.
J Gastroenterol ; 43(2): 136-43, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18306987

RESUMO

BACKGROUND: Erythropoietin (Epo) is the central regulator of red blood cell production and can stimulate proliferation and differentiation of erythroid progenitor cells. Now, recombinant human erythropoietin (rHuEpo) is widely used in patients with renal disease, chronic anemia, and iron deficiency of early childhood. It has been reported that the enhanced erythropoiesis associated with erythropoietin therapy increases intestinal iron absorption, but the molecular mechanisms underlying are unknown. Therefore, we have investigated the effect of rHuEpo on duodenal iron transport protein synthesis in rats. METHODS: Male Sprague-Dawley rats weighing 250 g were randomly divided into two groups: (1) rHuEpo injection group (rHuEpo, 500 IU/day, s.c.), and (2) control group (injection of the same volume of saline). After 3 days injection, blood parameters, serum iron status, and non-heme iron concentrations in the liver and duodenum were examined at the fifth day. The mRNA levels and protein synthesis of duodenal divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), and hephaestin (Hp) were measured by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis. Hepatic hepcidin mRNA expression was analyzed by RT-PCR. RESULTS: rHuEpo injection significantly stimulated erythropoiesis and decreased serum iron status, non-heme iron concentrations in the liver and duodenum. DMT1 (+IRE) and Hp expression in duodenum were increased significantly. However, DMT1 (-IRE) and FPN1 expression had no apparent change. Hepatic hepcidin mRNA expression was decreased dramatically, reaching an almost undetectable level in rHuEpo-treated rats. CONCLUSIONS: rHuEpo administration improved the duodenal iron absorption by increasing the expression of DMT1 (+IRE) and Hp.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Eritropoetina/farmacologia , Hematínicos/farmacologia , Absorção Intestinal/efeitos dos fármacos , Proteínas Reguladoras de Ferro/metabolismo , Proteínas de Membrana/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Western Blotting , Proteínas de Transporte de Cátions/genética , Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hepcidinas , Imuno-Histoquímica , Proteínas Reguladoras de Ferro/genética , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...