Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 13: 976117, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36188586

RESUMO

Background: Melanoma is the most lethal skin malignant tumor with a short survival once stepping into the metastatic status and poses a therapeutic challenge. Apatinib (a tyrosine kinase inhibitor) is a promising antiangiogenic agent for the treatment of metastatic melanoma. However, antiangiogenic monotherapy is prone to acquired drug resistance and has a limited therapeutic effect. The persistence dependence of glycolytic metabolism in antiangiogenic therapy-resistant cells provides evidence that glycolysis inhibitors may enhance the effect of antiangiogenic therapy. So, this study aimed to investigate whether WZB117 (a specific GLUT1 inhibitor) could enhance the anti-tumor effect of apatinib against melanoma and its potential mechanisms. Methods: We investigated the anti-tumor effects of apatinib alone or in combination with WZB117 on human melanoma cell lines (A375 and SK-MEL-28). The MTT assay determined cell viability and the half-maximal inhibitory concentration (IC50). Multiple drug effect/combination indexes (CI) analysis was conducted to assess interactions between apatinib and WZB117. Signal transducer and activator of transcription 3 (STAT3) pathway measured by western blotting and immunofluorescence staining. RNA expression analyses were performed using the reverse transcription-quantitative PCR method. Results: Apatinib and WZB117 showed dose and time-dependent growth inhibitory effects in both melanoma cells. The IC50 of apatinib at 48 h in A375 and SK-MEL-28 cells was 62.58 and 59.61 µM, respectively, while the IC50 of WZB117 was 116.85 and 113.91 µM, respectively. The CI values of the two drugs were 0.538 and 0.544, respectively, indicating a synergistic effect of apatinib combined with WZB117. We also found that glucose consumption and lactate production were suppressed by apatinib plus WZB117 in a dose-dependent manner, paralleled by reducing glycolytic enzyme pyruvate kinase M2 (PKM2). The potential mechanism of the combination was to suppress the phosphorylation of STAT3. Knockdown of STAT3 by siRNA inhibited the expression of PKM2, while the activation of STAT3 by IL-6 increased the expression of PKM2. The effects of IL-6 were attenuated by apatinib combined with WZB117 treatment. Conclusion: WZB117 enhanced the anti-tumor effect of apatinib against melanoma via modulating glycolysis by blocking the STAT3/PKM2 axis, which suggested the combination of apatinib with WZB117 could be a potential therapeutic candidate for melanoma.

2.
Neuron ; 110(18): 2949-2960.e4, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-35931070

RESUMO

Transmission from striatal cholinergic interneurons (CINs) controls dopamine release through nicotinic acetylcholine receptors (nAChRs) on dopaminergic axons. Anatomical studies suggest that cholinergic terminals signal predominantly through non-synaptic volume transmission. However, the influence of cholinergic transmission on electrical signaling in axons remains unclear. We examined axo-axonal transmission from CINs onto dopaminergic axons using perforated-patch recordings, which revealed rapid spontaneous EPSPs with properties characteristic of fast synapses. Pharmacology showed that axonal EPSPs (axEPSPs) were mediated primarily by high-affinity α6-containing receptors. Remarkably, axEPSPs triggered spontaneous action potentials, suggesting that these axons perform integration to convert synaptic input into spiking, a function associated with somatodendritic compartments. We investigated the cross-species validity of cholinergic axo-axonal transmission by recording dopaminergic axons in macaque putamen and found similar axEPSPs. Thus, we reveal that synaptic-like neurotransmission underlies cholinergic signaling onto dopaminergic axons, supporting the idea that striatal dopamine release can occur independently of somatic firing to provide distinct signaling.


Assuntos
Dopamina , Receptores Nicotínicos , Axônios/metabolismo , Colinérgicos , Fibras Colinérgicas/metabolismo , Corpo Estriado/fisiologia , Dopamina/fisiologia , Interneurônios/metabolismo , Receptores Nicotínicos/metabolismo , Transmissão Sináptica/fisiologia
3.
Cancer Med ; 11(11): 2271-2283, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35191609

RESUMO

BACKGROUND: The ALTER0203 clinical trial showed that anlotinib, a multitargeted tyrosine kinase inhibitor, had antitumor effects on advanced soft tissue sarcoma (STS) after the failure of standard chemotherapy. We aimed to evaluate the real-world efficacy and explore prognostic factors and treatment patterns of anlotinib in patients with advanced STS. METHODS: We retrospectively analyzed the data of patients with unresectable locally advanced or metastatic STS who received at least one dose of anlotinib from June 2018 to March 2021. The survival data were analyzed using the Kaplan-Meier method and compared using the log-rank test. The Cox proportional hazards model was performed for multivariate analysis. RESULTS: A total of 209 patients were included. The median age was 48 (range 11-85) years. The median follow-up, progression-free survival, and overall survival were 18.7 months, 6.1 months [95% confidence interval (CI): 4.9-7.2], and 16.4 months (95% CI: 13.6-19.1), respectively. The objective response rate was 13.4%. Nutritional status, Eastern Cooperative Oncology Group (ECOG) performance status, and anlotinib treatment patterns (combination therapy or switch maintenance therapy vs. monotherapy) were significantly associated with progression-free survival. Besides, pathological grade, nutritional status, ECOG performance status, and anlotinib treatment patterns were predictive of overall survival. Due to anlotinib-related toxicity, 31 (14.8%) patients, and 25 (12.0%) patients experienced dose reduction and treatment discontinuation, respectively. CONCLUSION: These findings confirmed the efficacy of anlotinib in patients with advanced STS in a real-world setting. The patterns of anlotinib treatment deserve further exploration.


Assuntos
Segunda Neoplasia Primária , Quinolinas , Sarcoma , Neoplasias de Tecidos Moles , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Humanos , Indóis , Pessoa de Meia-Idade , Segunda Neoplasia Primária/tratamento farmacológico , Quinolinas/efeitos adversos , Estudos Retrospectivos , Sarcoma/patologia , Adulto Jovem
4.
Cell Rep ; 32(11): 108156, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32937133

RESUMO

Substantia nigra (SNc) dopaminergic neurons respond to aversive stimuli with inhibitory pauses in firing followed by transient rebound activation. We tested integration of inhibitory synaptic inputs onto SNc neurons from genetically defined populations in dorsal striatum (striosome and matrix) and external globus pallidus (GPe; parvalbumin- and Lhx6-positive), and examined their contribution to pause-rebound firing. Activation of striosome projections, which target "dendron bouquets" in the pars reticulata (SNr), consistently quiets firing and relief from striosome inhibition triggers rebound activity. Striosomal inhibitory postsynaptic currents (IPSCs) display a prominent GABA-B receptor-mediated component that strengthens the impact of SNr dendrite synapses on somatic excitability and enables rebounding. By contrast, GPe projections activate GABA-A receptors on the soma and proximal dendrites but do not result in rebounding. Lastly, optical mapping shows that dorsal striatum selectively inhibits the ventral population of SNc neurons, which are intrinsically capable of rebounding. Therefore, we define a distinct striatonigral circuit for generating dopamine rebound.


Assuntos
Gânglios da Base/fisiologia , Neurônios Dopaminérgicos/fisiologia , Inibição Neural/fisiologia , Substância Negra/fisiologia , Animais , Cálcio/metabolismo , Corpo Estriado/fisiologia , Dendritos/fisiologia , Dopamina/metabolismo , Feminino , Globo Pálido/fisiologia , Masculino , Camundongos , Modelos Neurológicos , Receptores de GABA-A/metabolismo , Receptores de GABA-B/metabolismo , Sinapses/metabolismo
5.
Elife ; 92020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32870779

RESUMO

Axons of dopaminergic neurons innervate the striatum where they contribute to movement and reinforcement learning. Past work has shown that striatal GABA tonically inhibits dopamine release, but whether GABA-A receptors directly modulate transmission or act indirectly through circuit elements is unresolved. Here, we use whole-cell and perforated-patch recordings to test for GABA-A receptors on the main dopaminergic neuron axons and branching processes within the striatum of adult mice. Application of GABA depolarized axons, but also decreased the amplitude of axonal spikes, limited propagation and reduced striatal dopamine release. The mechanism of inhibition involved sodium channel inactivation and shunting. Lastly, we show the positive allosteric modulator diazepam enhanced GABA-A currents on dopaminergic axons and directly inhibited release, but also likely acts by reducing excitation from cholinergic interneurons. Thus, we reveal the mechanisms of GABA-A receptor modulation of dopamine release and provide new insights into the actions of benzodiazepines within the striatum.


Assuntos
Corpo Estriado/fisiologia , Diazepam/farmacologia , Inibição Neural , Receptores de GABA-A , Animais , Benzodiazepinas/farmacologia , Neurônios Colinérgicos/efeitos dos fármacos , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Camundongos , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Canais de Sódio/metabolismo , Ácido gama-Aminobutírico/metabolismo
6.
Biochem Pharmacol ; 180: 114108, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32569628

RESUMO

Noninfectious (autoimmune and immune-mediated) uveitis is an ocular inflammatory disease which can lead to blindness in severe cases. Due to the potential side effects of first-line drugs for clinical uveitis, novel drugs and targets against uveitis are still urgently needed. In the present study, using rat experimental autoimmune uveitis (EAU) model, we first found that minocycline treatment can substantially inhibit the development of EAU and improve the retinal function by suppressing the retinal microglial activation, and block the infiltration of inflammatory cells, including Th17, into the retina by decreasing the major histocompatibility complex class II (MHC II) expression in resident and infiltrating cells. Moreover, we demonstrated that minocycline treatment can remodel the gut microenvironment of EAU rats by restoring the relative abundance of Ruminococcus bromii, Streptococcus hyointestinalis, and Desulfovibrio sp. ABHU2SB and promoting a functional shift in the gut via reversing the levels of L-proline, allicin, aceturic acid, xanthine, and leukotriene B4, and especially increasing the production of propionic acid, histamine, and pantothenic acid. At last, we revealed that minocycline treatment can significantly attenuate the progression of EAU after inflammation onset, which may be explained by the role of minocycline in the remodeling of the gut microenvironment since selective elimination of retinal microglia on the later stages of EAU was shown to have little effect. These data clearly demonstrated that inhibition of microglial activation and remodeling of the gut microenvironment can suppress the development and progression of experimental autoimmune uveitis. Considering the excellent safety profile of minocycline in multiple clinical experiments, we suggest that minocycline may have therapeutic implications for clinical uveitis.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Microglia/efeitos dos fármacos , Minociclina/uso terapêutico , Retina/efeitos dos fármacos , Uveíte/tratamento farmacológico , Animais , Doenças Autoimunes/imunologia , Microambiente Celular/efeitos dos fármacos , Microambiente Celular/imunologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/imunologia , Antígenos de Histocompatibilidade Classe II/biossíntese , Masculino , Microglia/imunologia , Ratos , Ratos Endogâmicos Lew , Retina/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Células Th17/patologia , Uveíte/imunologia , Uveíte/patologia
7.
J Control Release ; 317: 109-117, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31778740

RESUMO

Subcellular delivery of nanomedicines has emerged as a promising approach to enhance the therapeutic efficacy of anticancer drugs. Nuclear accumulation of anticancer drugs are essential for its therapeutic efficacy because their targets are generally located within the nucleus. However, strategies for the nuclear accumulation of nanomedicines with anticancer drugs rarely reported. In this study, we reported a promising nanomedicine, comprising a drug-peptide amphiphile, with enhanced cellular uptake and nuclear accumulation capability for cancer therapy. The drug-peptide amphiphile consisted of the peptide ligand PMI (TSFAEYWNLLSP), which was capable of activating the p53 gene by binding with the MDM2 and MDMX located in the cell nucleus. Peptide conformations could be finely tuned by using different strategies including heating-cooling and enzyme-instructed self-assembly (EISA) to trigger molecular self-assembly at different temperatures. Due to the different peptide conformations, the drug-peptide amphiphile self-assembled into nanomedicines with various properties, including stabilities, cellular uptake, and nuclear accumulation. The optimized nanomedicine formed by EISA strategy at a low temperature of 4 °C showed enhanced cellular uptake and nuclear accumulation capability, and thus exhibited superior anticancer ability both in vitro and in vivo. Overall, our study provides a useful strategy for finely tuning the properties and activities of peptide-based supramolecular nanomaterials, which may lead to optimized nanomedicines with enhanced performance.


Assuntos
Antineoplásicos , Nanoestruturas , Preparações Farmacêuticas , Nanomedicina , Peptídeos
8.
Chem Commun (Camb) ; 55(35): 5123-5126, 2019 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-30973557

RESUMO

Supramolecular hydrogels of self-assembling peptides and thermogels are very promising for biomedical applications. However, there were no thermogels of self-assembling peptides. In this study, we reported on a novel and versatile strategy to prepare thermogels of self-assembling peptides by enzyme-instructed peptide folding and self-assembly. We synthesized two phosphorylated peptides from insulin growth factor (IGF) and the second mitochondria-derived activator of caspases (Smac) (Nap-FFGGpYGSSSRRAPQT and NBD-GFFpYGAVPIAQK, respectively), which could be converted to possible hydrogelators by enzyme-instructed self-assembly (EISA). We found that EISA using phosphatase at 4 °C resulted in peptides with a random coil conformation, which would self-assemble into worm-like micelles or very short fibers in clear solutions. At a physiological temperature of 37 °C, the peptides would undergo fast transitions from random coil to ß-sheet- or α-helix-like conformations, resulting in solution-to-gel transformations. This novel thermogel system was very useful for three-dimensional (3D) cell culture due to the biocompatibility and bioactivity of peptides. Our study provides a novel strategy to prepare a novel thermogel system for biomedical applications.


Assuntos
Hidrogéis/química , Fragmentos de Peptídeos/química , Fosfopeptídeos/química , Fosfatase Alcalina/química , Sequência de Aminoácidos , Animais , Técnicas de Cultura de Células/métodos , Hidrogéis/síntese química , Hidrólise , Fator de Crescimento Insulin-Like I/química , Células-Tronco Mesenquimais/metabolismo , Proteínas Mitocondriais/química , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Dobramento de Proteína , Multimerização Proteica , Ratos , Temperatura de Transição
9.
J Biomed Nanotechnol ; 15(4): 822-829, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30841974

RESUMO

Topical ocular drug administration aimed at providing a high drug concentration at the precorneal site accompanied with enhanced corneal permeability to avoid systemic side effects is a very important therapeutic goal in ocular disorder therapy. In the present study, the solubility of the poorly soluble drug celecoxib (CXB) was significantly improved using a facile strategy to generate a high drug payload micellar formulation. By varying the drug/polymer feed ratios, the mean diameter of the formed CXB micelles ranged from 21.34 ± 0.23 to 28.53 ± 0.11 nm, and the drug loading capacity ranged from 4.31 to 15.87%. Transmission electron microscopy (TEM) showed that the formed CXB micelles had a uniform spherical morphology. Differential scanning calorimetry (DSC), X-ray powder diffraction (XRD) and Fourier transform infrared spectroscopy (FTIR) were used to characterize the physicochemical properties of lyophilized CXB micelles. The obtained CXB micelles retained their properties through freeze-drying and rehydration, thereby providing long-term physicochemical stability over 3 months of storage at -20 °C. An in vitro release study showed that the CXB micelles released CXB in a sustained release manner without any apparent burst release over 72 h by the Higuchi non-Fickian diffusion mechanism. Notably, using corneas excised from rabbits, the in vitro corneal permeability of CXB from the micellar formulation was observed to be significantly greater than that of the microparticle formulation. Overall, the proposed micelles might be a promising vehicle for ophthalmic delivery of CXB with the significant enhancement of water solubility and corneal permeability.


Assuntos
Córnea , Animais , Varredura Diferencial de Calorimetria , Celecoxib , Micelas , Permeabilidade , Coelhos , Solubilidade
10.
Int J Nanomedicine ; 14: 591-603, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30666116

RESUMO

PURPOSE: To develop and demonstrate the effectiveness of a novel dexamethasone (Dex) nanoformulation for treating uveitis. MATERIALS AND METHODS: We designed and screened a dexamethasone-peptide conjugate (Dex-SA-FFFE), formed via a biodegradable ester bond linkage, that could spontaneously form high drug payload nanoparticles in aqueous solution for treating uveitis. RESULTS: An in vitro release study indicated that Dex and Dex-SA-FFFE sustainably released from Dex-SA-FFFE nanoparticles over a 48 h study period. Meanwhile, the formed Dex-SA-FFFE nanoparticles hardly caused cytotoxicity in human corneal epithelial cell at drug concentrations up to 1 mM after 24 h of incubation but reduced cell viability after 48 h and 72 h of incubation. An in vitro anti-inflammatory efficacy assay showed that the Dex-SA-FFFE nanoparticles exhibited a comparable anti-inflammatory efficacy to that of Dex in lipopolysaccharide (LPS)-activated RAW264.7 macrophages via significant decreases in the secretion of various pro-inflammatory cytokines (e.g., nitric oxide, tumor necrosis factor-α, interleukin-6). Topical instillation of Dex-SA-FFFE nanoparticles showed good ocular tolerance without causing changes in corneal thickness and intraocular pressure during the entire study period. Furthermore, topical instillation of Dex-SA-FFFE nanoparticles displayed a comparable in vivo therapeutic efficacy to that of dexamethasone sodium phosphate (Dexp) aqueous solutions in an endotoxin-induced uveitis (EIU) rabbit model. CONCLUSION: Based on these results, it is reasonable to believe that the proposed Dex-SA-FFFE nanoparticles might have great application for the treatment of anterior uveitis.


Assuntos
Dexametasona/uso terapêutico , Nanopartículas/química , Peptídeos/química , Uveíte/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Morte Celular/efeitos dos fármacos , Dexametasona/farmacologia , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Olho/efeitos dos fármacos , Humanos , Interleucina-6/biossíntese , Contagem de Leucócitos , Lipopolissacarídeos , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Óxido Nítrico/biossíntese , Tamanho da Partícula , Células RAW 264.7 , Coelhos , Ácido Succínico/química , Fator de Necrose Tumoral alfa/biossíntese , Uveíte/sangue , Uveíte/patologia
11.
Research (Wash D C) ; 2019: 4803624, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31912037

RESUMO

The selective formation of nanomaterials in cancer cells and tumors holds great promise for cancer diagnostics and therapy. Until now, most strategies rely on a single trigger to control the formation of nanomaterials in situ. The combination of two or more triggers may provide for more sophisticated means of manipulation. In this study, we rationally designed a molecule (Comp. 1) capable of responding to two enzymes, alkaline phosphatase (ALP), and reductase. Since the A549 lung cancer cell line showed elevated levels of extracellular ALP and intracellular reductase, we demonstrated that Comp. 1 responded in a stepwise fashion to those two enzymes and displayed a tandem molecular self-assembly behavior. The selective formation of nanofibers in the mitochondria of the lung cancer cells led to the disruption of the mitochondrial membrane, resulting in an increased level of reactive oxygen species (ROS) and the release of cytochrome C (Cyt C). ROS can react with proteins, resulting in endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). This severe ER stress led to disruption of the ER, formation of vacuoles, and ultimately, apoptosis of the A549 cells. Therefore, Comp. 1 could selectively inhibit lung cancer cells in vitro and A549 xenograft tumors in vivo. Our study provides a novel strategy for the selective formation of nanomaterials in lung cancer cells, which is powerful and promising for the diagnosis and treatment of lung cancer.

12.
Colloids Surf B Biointerfaces ; 175: 569-575, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30580147

RESUMO

Combining a low-molecular-weight hydrogel (LMWH) with a polymeric hydrogel overcomes the disadvantages of the LMWH (e.g., its low mechanical property) and is associated with the enhancement of materials performance, which is useful in a variety of biomedical applications. In the present work, a hybrid hydrogel that combines dexamethasone sodium phosphate (Dexp) and a polysaccharide (alginate) was explored via a calcium ion coordination strategy. With the addition of Ca2+ to an aqueous solution of Dexp/alginate, the Ca2+/Dexp/alginate hybrid hydrogel formed spontaneously. The formed Ca2+/Dexp/alginate hybrid hydrogels were thoroughly characterized by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR) and powder X-ray diffraction (XRD). An in vitro drug release study indicated that the formed Ca2+/Dexp/alginate hybrid hydrogel provided a slower drug release rate than did the Ca2+/Dexp hydrogel, and the drug release behaviour could be finely tailored by the change of Ca2+ concentration. More importantly, the subcutaneous injection of the Ca2+/Dexp/alginate hybrid hydrogel significantly extended the in vivo retention of the hydrogel in situ compared to that of the Ca2+/Dexp hydrogel. The in vivo pharmacokinetic analysis indicated that the Ca2+/Dexp/alginate hybrid hydrogel could greatly extend drug release in vivo and significantly improve drug bioavailability compared to the Ca2+/Dexp hydrogel. As such, the formed Ca2+/Dexp/alginate hybrid hydrogel combined the greater resilience of an alginate network with the long in vivo duration of a low-molecular-weight hydrogel (Ca2+/Dexp hydrogel) and remarkably enhanced drug bioavailability, which might open an avenue for the design of self-assembling steroidal drug-polysaccharide hybrid hydrogels for drug delivery applications.


Assuntos
Alginatos/química , Cloreto de Cálcio/química , Preparações de Ação Retardada/síntese química , Dexametasona/análogos & derivados , Portadores de Fármacos/síntese química , Hidrogéis/química , Animais , Dexametasona/sangue , Dexametasona/química , Dexametasona/farmacocinética , Liberação Controlada de Fármacos , Injeções Subcutâneas , Cinética , Camundongos , Camundongos Endogâmicos BALB C
13.
Int J Pharm ; 551(1-2): 223-231, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30213683

RESUMO

In this paper, we presented a simple yet versatile strategy to generate a high drug payload nanoparticles by the combination with small molecular assembly and polymeric assembly for topical suppression of ocular inflammation. Upon physical mixing of the succinated triamcinolone acetonide (TA-SA) supramolecular hydrogel with the poly (ethylene glycol)-poly (ɛ-caprolactone)-poly (ethylene glycol) (PECE) aqueous solution at 37 °C, TA-SA/PECE nanoparticles formed spontaneously and characterized thoroughly by transmission electron microscopy (TEM), X-ray diffraction (XRD) and differential scanning calorimetry (DSC). The formed TA-SA/PECE nanoparticles displayed a comparable in vitro anti-inflammatory efficacy to that of native triamcinolone acetonide (TA), through a significant downregulation of various proinflammatory cytokines levels (e.g., NO, TNF-α) in a lipopolysaccharide (LPS) actived RAW264.7 macrophage. Meanwhile, the enhanced transcorneal drug permeability of TA-SA/PECE nanoparticles over that of TA suspension was clearly observed in an isolated rabbit cornea. Intraocular biocompatibility test demonstrated that TA-SA/PECE nanoparticles presented good biocompatibility after topical instillation during entire study period. More importantly, the TA-SA/PECE nanoparticles displayed superior therapeutic efficacy over that of the TA suspension in the endotoxin-induced uveitis (EIU) rabbit model via decreasing neutrophil infiltration in anterior chamber. Overall, the proposed TA-SA/PECE nanoparticles might be a promising candidate for uveitis therapy.


Assuntos
Anti-Inflamatórios/administração & dosagem , Hidrogéis/administração & dosagem , Nanopartículas/administração & dosagem , Poliésteres/administração & dosagem , Polietilenoglicóis/administração & dosagem , Triancinolona Acetonida/administração & dosagem , Uveíte/tratamento farmacológico , Administração Tópica , Animais , Linhagem Celular , Córnea/metabolismo , Humanos , Lipopolissacarídeos , Camundongos , Polímeros/administração & dosagem , Coelhos , Uveíte/induzido quimicamente
14.
J Biomed Nanotechnol ; 14(8): 1471-1476, 2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-29903061

RESUMO

In the present study, we reported the generation of a molecular hydrogel of succinated paclitaxel (PTX-SA) by a self-hydrolytic strategy for potential local cancer chemotherapy. Upon self-hydrolysis of the ester bond of PTX-SA in phosphate-buffered saline (pH = 7.4) for 24 h, a PTX-SA supramolecular hydrogel formed spontaneously with a minimal gelation concentration of 0.25 wt%. The formed PTX-SA supramolecular hydrogel displayed a filamentous nanostructure, the nanofibers of which were typically several micrometers in length with a diameter of 10-15 nm. Rheological analysis suggested that the PTX-SA supramolecular hydrogel exhibited dominant elastic and thixotropic properties. Ionization of the carboxylate group together with the self-hydrolysis of the ester bond in PTX-SA are the major driving forces for the hydrogelation, as indicated by Fourier transform infrared spectroscopy (FTIR) analysis. The bioactive drug payload sustainably released from the supramolecular hydrogel over a period of 15 day in an in vitro release study, and the drug-release behavior could be finely controlled by altering the initial PTX-SA concentration. More importantly, the formed PTX-SA supramolecular hydrogel without compromising its pharmacological activity could efficiently inhibit the proliferation of cancer cells (HepG2 and MCF-7 cells) in vitro. Therefore, the generated PTX-SA supramolecular hydrogel might provide great potential as a novel drug-delivery system for local anti-cancer therapy.


Assuntos
Nanofibras , Paclitaxel/química , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Hidrogéis
15.
J Colloid Interface Sci ; 528: 10-17, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-29803956

RESUMO

In the present study, we propose an ionic coordination strategy for the design of a steroidal prodrug supramolecular hydrogel. The hydrogel composed of nanofibril networks formed spontaneously by the introduction of divalent cations (e.g., Mg2+, Ca2+, Zn2+ and Fe2+) and NH4+ to a succinated dexamethasone (Dex-SA) aqueous solution at room temperature. The formation of the nanofibril structure was dominantly driven by the ionic coordination with the assistance of a delicate balance of multiple noncovalent interactions. A rheological analysis indicated that the formed Ca2+/Dex-SA supramolecular hydrogel exhibits dominant elastic and thixotropic properties. The formed Ca2+/Dex-SA supramolecular hydrogel allowed the gradual release of Dex and Dex-SA in vitro, and the drug release behaviour can be finely tuned by changing the Ca2+ concentration. Storage stability studies showed that Dex-SA in hydrogel underwent an apparent chemical decomposition at 4 °C and 37 °C. In contrast, the Dex-SA xerogel was quite stable without any obvious chemical decomposition of Dex-SA in storage at -20 °C for 35 days, and it was able to turn into a hydrogel again within one minute after rehydration. The formed Ca2+/Dex-SA supramolecular hydrogel caused negligible cytotoxicity against HCEC and L-929 cells at drug concentrations up to 2 mM, as indicated by the in vitro cytotoxicity tests. Additionally, the proposed Ca2+/Dex-SA supramolecular hydrogel displayed a comparable anti-inflammatory efficacy with Dexp via the downregulation of NO, TNF-α and IL-6 expression in lipopolysaccharide (LPS)-activated RAW264.7 macrophage. Overall, the cation instructed steroidal prodrug supramolecular hydrogel might be a promising ophthalmic drug delivery system for anti-inflammatory therapy.


Assuntos
Anti-Inflamatórios/administração & dosagem , Cátions Bivalentes/química , Dexametasona/administração & dosagem , Hidrogéis/química , Nanofibras/química , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/farmacologia , Linhagem Celular , Dexametasona/química , Dexametasona/farmacocinética , Dexametasona/farmacologia , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Elasticidade , Humanos , Camundongos , Nanofibras/ultraestrutura , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Células RAW 264.7 , Ácido Succínico/administração & dosagem , Ácido Succínico/química , Ácido Succínico/farmacocinética , Ácido Succínico/farmacologia
16.
Acta Biomater ; 73: 275-284, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29660509

RESUMO

Intravitreal/periocular injection of triamcinolone acetonide (TA) suspension is a common uveitis treatment, but it displays a high risk for serious side effects (e.g., high intraocular pressure, retinal toxicity). We report here an intravitreally injectable thermosensitive glycosylated TA (TA-SA-Glu) hydrogel, formed by covalently conjugating glucosamine (Glu) with succinate TA (TA-SA), for treating uveitis. The TA-SA-Glu hydrogelator forms a supramolecular hydrogel spontaneously in aqueous solution with a minimal gelation concentration of 0.25 wt%. Structural analysis revealed that hydrogen bonds assisted by hydrophobic interaction resulted in self-assembled nanofibers. Rheology analysis demonstrated that this TA-SA-Glu hydrogel exhibited a typical thixotropic property. Sustained release of both TA-SA-Glu and TA from the hydrogel occurred throughout the 3-day in vitro release study. The obtained TA-SA-Glu hardly caused cytotoxicity against ARPE-19 and RAW264.7 cells after 24 h of incubation at drug concentration up to 600 µM. In particular, TA-SA-Glu exhibited a comparable anti-inflammatory efficacy to TA in terms of inhibiting the production of nitric oxide, tumor necrosis factor-α, and interleukin-6 in activated RAW264.7 macrophages. Following a single intravitreal injection, 69 nmol TA-SA-Glu hydrogel caused minimal apparent retinal toxicity, whereas the TA suspension displayed significant effects in terms of localized retinal toxicity. A single intravitreal injection of TA-SA-Glu hydrogel was more effective in controlling inflammatory response than that of the TA suspension treatment, particularly in down-regulating the pro-inflammatory Th1 and Th17 effector responses for treating experimental autoimmune uveitis. This study strongly indicates that supramolecular TA-SA-Glu hydrogels may represent a new option for posterior uveitis management. STATEMENT OF SIGNIFICANCE: Intravitreal/periocular injection of triamcinolone acetonide (TA) suspension is a common uveitis treatment, but suffers a high risk for serious side effects (e.g., high intraocular pressure, retinal toxicity). We generated an injectable glycosylated triamcinolone acetonide hydrogelator (TA-SA-Glu) hydrogel for treating uveitis. Following a single intravitreal injection, the proposed TA-SA-Glu hydrogel hardly caused apparent retinal toxicity at a dosage of 69 nmol per eye. Furthermore, TA-SA-Glu hydrogel was more effective in controlling non-infectious uveitis over than a TA suspension, particularly in terms of down-regulating the pro-inflammatory Th1 and Th17 effector responses for treating experimental autoimmune uveitis (EAU). This study strongly indicates that TA-SA-Glu supramolecular hydrogels may represent a new option for the management of various intraocular inflammations.


Assuntos
Anti-Inflamatórios/farmacologia , Doenças Autoimunes/tratamento farmacológico , Hidrogéis/química , Inflamação/tratamento farmacológico , Triancinolona Acetonida/farmacologia , Uveíte/tratamento farmacológico , Administração Tópica , Animais , Anti-Inflamatórios/administração & dosagem , Materiais Biocompatíveis/química , Relação Dose-Resposta a Droga , Eletrorretinografia , Glucosamina/administração & dosagem , Glicosilação , Ligação de Hidrogênio , Injeções Intravítreas , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Células RAW 264.7 , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Retina/efeitos dos fármacos , Reologia , Espectroscopia de Infravermelho com Transformada de Fourier , Ácido Succínico/administração & dosagem , Células Th1/efeitos dos fármacos , Triancinolona Acetonida/administração & dosagem
17.
Colloids Surf B Biointerfaces ; 164: 436-443, 2018 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-29438842

RESUMO

The low bioavailability exhibits by conventional ocular formulation owing to rapid precorneal clearance and lower corneal permeability can be overcame by the application of the gelling system. In the present study, a prodrug supramolecular hydrogel derived from succinated dexamethasone (Dex-SA) was fabricated using a pH hydrolytic strategy and explored as a "self-delivery" system for ophthalmic drugs. The self-assembled Dex-SA supramolecular hydrogel exhibited a typical nano-fibrous microstructure and was thixotropic. Both dexamethasone (Dex) and Dex-SA prodrug sustainably released from Dex-SA supramolecular hydrogel in a period of 120 h in vitro release study, and the initial pH value of hydrogel significantly influence on the release ratio of Dex/Dex-SA. Furthermore, the lyophilized Dex-SA supramolecular hydrogel displayed long-term stability without causing any apparent hydrolysis of Dex-SA at -20 °C over 30 day and quickly re-formed a hydrogel after dissolving into aqueous solution. The formed Dex-SA supramolecular hydrogel had lower cytotoxicity than Dex at drug concentration up to 2.5 mM, and exhibited a comparable anti-inflammatory efficacy to a Dex sodium phosphate (Dexp) aqueous solution in lipopolysaccharide-activated RAW264.7 macrophages. Topical instillation of the Dex-SA supramolecular hydrogel showed excellent intraocular biocompatibility and it was not an irritant in rabbit eyes. More importantly, the Dex-SA supramolecular hydrogel provided a prolonged precorneal retention and significantly enhanced the ocular bioavailability over Dexp aqueous solution after topical instillation. Overall, this work illustrates an effective approach for the development of prodrug supramolecular hydrogels to extend the precorneal retention and enhance ocular bioavailability of drugs after topical instillation.


Assuntos
Dexametasona/administração & dosagem , Dexametasona/farmacologia , Sistemas de Liberação de Medicamentos , Olho/efeitos dos fármacos , Hidrogéis/química , Nanofibras/química , Administração Tópica , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Dexametasona/farmacocinética , Liberação Controlada de Fármacos , Humanos , Irritantes , Masculino , Camundongos , Nanofibras/ultraestrutura , Coelhos , Espectroscopia de Infravermelho com Transformada de Fourier
18.
Am J Mens Health ; 11(1): 92-98, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25990510

RESUMO

The growth of prostate tumors is mediated by the bioavailability of androgens and insulin-like growth factors. This study tested the hypothesis that healthy young adult African American men exhibiting low aerobic capacity (fitness) have serum insulin-like growth Factor-1 (IGF-1) and testosterone levels that promote growth of prostate cancer cells. A cross-sectional data research design was used to study groups of 18- to 26-year-old healthy men exhibiting low and moderate aerobic fitness, based on their peak oxygen consumption (VO2peak). The individual serum levels of IGF-1, IGF-1 binding protein-3 (IGFBP-3), and testosterone were measured. In vitro growth of androgen-dependent LNCaP prostate tumor cells was measured after incubation in culture medium fortified with each subject's serum. Aerobic capacity was significantly greater in the moderate-fitness group than in the low-fitness group without an intergroup difference in body mass index. The serum IGF-1 concentration was significantly higher in the low-fitness group in the absence of an intergroup difference in serum testosterone. The serum IGFBP-3 concentration was significantly lower in the low-fitness group. Prostate tumor cell growth was significantly greater in the cultures incubated in media containing the sera of the low-fitness group than in the sera of the moderate-fitness group. These findings suggest that moderate aerobic fitness in young adults may decrease the circulating levels of free IGF-1 and lower the potential to support growth of prostate cancer cells.

19.
J Basic Clin Med ; 1(1): 16-19, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23745173

RESUMO

BACKGROUND: Hyperthermia is used in combination with radiotherapy and/or chemotherapy in the treatment of various types of cancer. Currently, the tumor cell response to hyperthermia is determined largely based on the size reduction of tumor mass, which is insensitive. METHODS: We tested the feasibility of bioluminescent imaging (BLI) in evaluation of the tumor cell response to hyperthermia by exposing luciferase-expressing MDA-MB-231-luc human breast cancer cells to high temperature (43 °C) for 10 minutes to 2 hours. The tumor cells were the imaged and the light signal generated by the tumor cells was quantified with BLI. To validate its usefulness, the light signal intensity was comparatively analyzed with the tumor cell clonogenicity and cell viability, which were measured with classic clonogenic and MTT assays. RESULTS: The light signal intensity determined by BLI was closely correlated with the absolute number of viable cells as well as the cell viability measured with the traditional MTT assay under normal culture condition. Relative to the clonogenicity of tumor cells after exposure to hyperthermia, however, BLI underestimated, while MTT assay overestimated the cell viability. Difference in the interpretation of tumor cell clonogenic ability following hyperthermia with BLI, MTT dye, and clonogenic assay may be due to the different mechanisms of the three measurements as well as the fact that hyperthermia can induce cell damage at levels of both transient and permanent. CONCLUSIONS: BLI is sensitive, convenient, and potentially valuable in the evaluation and monitoring of tumor cell response to treatments including hyperthermia.

20.
Anal Biochem ; 382(2): 122-8, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18722992

RESUMO

We have developed an image-based technique for signal pathway analysis, target validation, and compound screening related to mammary epithelial cell differentiation. This technique used the advantages of optical imaging and the HC11-Lux model system. The HC11-Lux cell line is a subclone of HC11 mammary epithelial cells transfected stably with a luciferase construct of the beta-casein gene promoter (p-344/-1betac-Lux). The promoter activity was imaged optically in real time following lactogenic induction. The imaging signal intensity was closely correlated with that measured using a luminometer following protein extraction (R=0.99, P<0.0001) and consistent with the messenger RNA (mRNA) level of the endogenous beta -casein gene. Using this technique, we examined the roles of JAK2/Stat5A, Raf-1/MEK/MAKP, and PI3K/Akt signal pathways with respect to differentiation. The imaging studies showed that treatment of the cells with epidermal growth factor (EGF), AG490 (JAK2-specific inhibitor), and LY294002 (PI3K-specific inhibitor) blocked lactogenic differentiation in a dose-dependent manner. PD98059 (MEK-specific inhibitor) could reverse EGF-mediated differentiation arrest. These results indicate that these pathways are essential in cell differentiation. This simple, sensitive, and reproducible technique permits visualization and real-time evaluation of the molecular events related to milk protein production. It can be adopted for high-throughput screening of small molecules for their effects on mammary epithelial cell growth, differentiation, and carcinogenesis.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Medições Luminescentes , Glândulas Mamárias Animais/citologia , Animais , Caseínas/genética , Caseínas/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Feminino , Janus Quinase 2/metabolismo , Glândulas Mamárias Animais/metabolismo , Camundongos , Microscopia de Fluorescência , Proteínas Proto-Oncogênicas c-raf/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA