Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Cancer Gene Ther ; 31(6): 894-903, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38418842

RESUMO

This study utilized Next-Generation Sequencing (NGS) to explore genetic determinants of survival duration in Glioblastoma Multiforme (GBM) patients. We categorized 30 primary GBM patients into two groups based on their survival periods: extended survival (over two years, N = 17) and abbreviated survival (under two years, N = 13). For identifying pathogenic or likely pathogenic variants, we leveraged the ClinVar database. The cohort, aged 23 to 66 (median: 53), included 17 patients in Group A (survival >2 years, 10 males, 7 females), and 13 patients in Group B (survival <2 years, 8 males, 5 females), with a 60% to 40% male-to-female ratio. Identified mutations included CHEK2 (c.1477 G > A, p.E493K), IDH1 (c.395 G > A, p.R132H), and TP53 mutations. Non-coding regions exhibited variants in the TERT promoter (c.-146C > T, c.-124C > T) and TP53 RNA splicing site (c.376-2 A > C, c.376-2 A > G). While Group A had more mutations, statistical significance wasn't reached, likely due to sample size. Notably, TP53, and ATR displayed a trend toward significance. Surprisingly, TP53 mutations were more prevalent in Group A, contradicting Western findings on poorer GBM prognosis. In Taiwanese GBM patients, bevacizumab usage is linked to improved survival rates, affirming its safety and effectiveness. EGFR mutations are infrequent, suggesting potential distinctions in carcinogenic pathways. Further research on EGFR mutations and amplifications is essential for refining therapeutic approaches. TP53 mutations are associated with enhanced survival, but their functional implications necessitate detailed exploration. This study pioneers genetic analysis in Taiwanese GBM patients using NGS, advancing our understanding of their genetic landscape.


Assuntos
Glioblastoma , Mutação , Humanos , Glioblastoma/genética , Glioblastoma/mortalidade , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Taiwan/epidemiologia , Idoso , Estudos Retrospectivos , Adulto Jovem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Sequenciamento de Nucleotídeos em Larga Escala , Prognóstico
2.
Biomater Adv ; 146: 213282, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36634378

RESUMO

Multidrug resistance (MDR) has been considered as a major adversary in oncologic chemotherapy. To simultaneously overcome drug resistance and inhibit tumor growth, it is essential to develop a drug delivery system that can carry and release multiple therapeutic agents with spatiotemporal control. In this study, we developed a hydrogel containing an enzyme-cleavable peptide motif, with a network structure formed by 4-armed polyethylene glycol (PEG) crosslinked by complementary nucleic acid sequences. Hydrogen bond formation between nucleobase pairing allows the hydrogel to be injectable, and the peptide motif grants deliberate control over hydrogel degradation and the responsive drug release. Moreover, MDR-targeted siRNAs are complexed with stearyl-octaarginine (STR-R8), while doxorubicin (Dox) is intercalated with DNA and nanoclay structures in this hydrogel to enhance therapeutic efficacy and overcome MDR. The results show a successful configuration of a hydrogel network with in situ gelation property, injectability, and degradability in the presence of tumor-associated enzyme, MMP-2. The synergistic effect by combining MDR-targeted siRNAs and Dox manifests with the enhanced anti-cancer effect on drug resistant breast cancer cells in both in vitro and in vivo tumor models. We suggest that with the tailor-designed hydrogel system, multidrug resistance in tumor cells can be significantly inhibited by the co-delivery of multiple therapeutics with spatial-temporal control release.


Assuntos
Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Hidrogéis , Neoplasias , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Hidrogéis/farmacologia , Neoplasias/tratamento farmacológico , RNA Interferente Pequeno , Humanos
3.
ACS Appl Mater Interfaces ; 15(1): 158-181, 2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35544684

RESUMO

Glioblastoma (GBM), classified as a grade IV glioma, is a rapidly growing, aggressive, and most commonly occurring tumor of the central nervous system. Despite the therapeutic advances, it carries an ominous prognosis, with a median survival of 14.6 months after diagnosis. Accumulating evidence suggests that cancer stem cells in GBM, termed glioma stem cells (GSCs), play a crucial role in tumor propagation, treatment resistance, and tumor recurrence. GSCs, possessing the capacity for self-renewal and multilineage differentiation, are responsible for tumor growth and heterogeneity, leading to primary obstacles to current cancer therapy. In this respect, increasing efforts have been devoted to the development of anti-GSC strategies based on targeting GSC surface markers, blockage of essential signaling pathways of GSCs, and manipulating the tumor microenvironment (GSC niches). In this review, we will discuss the research knowledge regarding GSC-based therapy and the underlying mechanisms for the treatment of GBM. Given the rapid progression in nanotechnology, innovative nanomedicines developed for GSC targeting will also be highlighted from the perspective of rationale, advantages, and limitations. The goal of this review is to provide broader understanding and key considerations toward the future direction of GSC-based nanotheranostics to fight against GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Neoplasias Encefálicas/patologia , Nanomedicina , Glioma/patologia , Glioblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
4.
J Colloid Interface Sci ; 628(Pt A): 717-725, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-35944302

RESUMO

Most biochemical reactions that occur in living organisms are catalyzed by a series of enzymes and proceed in a tightly controlled manner. The development of artificial enzyme cascades that resemble multienzyme complexes in nature is of current interest due to their potential in various applications. In this study, a nanozyme based on photoswitchable carbon-dot liposomes (CDsomes) was developed for use in programmable catalytic cascade reactions. These CDsomes prepared from triolein are amphiphilic and self-assemble into liposome-like structures in an aqueous environment. CDsomes feature excitation-dependent photoluminescence and, notably, can undergo reversible switching between a fluorescent on-state and nonfluorescent off-state under different wavelengths of light irradiation. This switching ability enables the CDsomes to exert photocatalytic oxidase- and peroxidase-like activities in their on- (bright) and off- (dark) states, respectively, resulting in the conversion of oxygen molecules into hydrogen peroxide (H2O2), followed by the generation of active hydroxyl radicals (OH). The two steps of oxygen activation can be precisely controlled in a sequential manner by photoirradiation at different wavelengths. Catalytic reversibility also enables the CDsomes to produce sufficient reactive oxygen species (ROS) to effectively kill tumor cells. Our results reveal that CDsomes is a promising photo-cycling nanozyme for precise tumor phototherapy through regulated programmable cascade reactions.


Assuntos
Peróxido de Hidrogênio , Lipossomos , Carbono , Catálise , Complexos Multienzimáticos/química , Oxirredutases , Oxigênio , Peroxidases , Espécies Reativas de Oxigênio , Trioleína
5.
J Nanobiotechnology ; 20(1): 235, 2022 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-35590324

RESUMO

BACKGROUND: MXenes with interesting optical and electrical properties have been attractive in biomedical applications such as antibacterial and anticancer agents, but their low photogeneration efficiency of reactive oxygen species (ROS) and poor stability are major concerns against microbial resistance. METHODS: Water-dispersible single layer Ti3C2Tx-based MXene through etching tightly stacked MAX phase precursor using a minimally intensive layer delamination method. After addition of Cu(II) ions, the adsorbed Cu(II) ions underwent self-redox reactions with the surface oxygenated moieties of MXene, leading to in situ formation of Cu2O species to yield Cu2O/Ti3C2Tx nanosheets (heterostructures). RESULTS: Under NIR irradiation, the Cu2O enhanced generation of electron-hole pairs, which boosted the photocatalytic production of superoxide and subsequent transformation into hydrogen peroxide. Broad-spectrum antimicrobial performance of Cu2O/Ti3C2Tx nanosheets with sharp edges is attributed to the direct contact-induced membrane disruption, localized photothermal therapy, and in situ generated cytotoxic free radicals. The minimum inhibitory concentration of Cu2O/Ti3C2Tx nanosheets reduced at least tenfold upon NIR laser irradiation compared to pristine Cu2O/Ti3C2Tx nanosheets. The Cu2O/Ti3C2Tx nanosheets were topically administrated on the methicillin-resistant Staphylococcus aureus (MRSA) infected wounds on diabetic mice. CONCLUSION: Upon NIR illumination, Cu2O/Ti3C2Tx nanosheets eradicated MRSA and their associated biofilm to promote wound healing. The Cu2O/Ti3C2Tx nanosheets with superior catalytic and photothermal properties have a great scope as an effective antimicrobial modality for the treatment of infected wounds.


Assuntos
Diabetes Mellitus Experimental , Staphylococcus aureus Resistente à Meticilina , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Camundongos , Oxirredução , Titânio/farmacologia
6.
J Chin Med Assoc ; 85(2): 198-203, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35175243

RESUMO

BACKGROUND: Trigeminal neuralgia (TN) is a disease characterized by recurring, short-lived, electric shock-like pain experienced on one side of the face. Microvascular decompression (MVD) is one of the most effective surgical interventions for resolving TN caused by neurovascular compression. This study aimed to determine the predictive and prognostic factors of surgical outcomes. METHODS: This retrospective cohort study enrolled patients diagnosed with TN who underwent MVD at our hospital during 2013-2019. The demographic information, pain character, peri-operative Barrow Neurological Institute (BNI) scale, medication, operative finding were recorded. And the outcome was Outcomes were divided into drug-free and drug-dependent group. Predisposing factors for each outcome were analyzed by one-way analysis of variance, followed by a Mann-Whitney U test or Kruskal-Wallis test. RESULTS: A total of 104 consecutive patients received MVD to treat TN, and 88 patients were enrolled in this study. The overall postoperative drug-free outcome was 72.7%. A significant difference in drug-free outcomes was observed for patients with typical TN (80.8%) compared with patients with atypical TN (33.33%, p = 0001). When severe venous compression was encountered during MVD, the drug-free outcome fell to 50% (10/20, p = 0.009). The Mann-Whitney U test indicated typical TN as a positive predictive factor of a drug-free outcome, whereas severe venous compression was a negative predictive factor. The patients with preoperative BNI score of 4 had better improvement than others (p = 0.045). Age, onset duration, and arterial loop had no specific difference in this study. CONCLUSION: In our study, atypical TN and severe venous compression were associated with poor outcomes. Regrouping atypical TN into precise diagnosis represents an immediate priority according to our result. The preoperative BNI score could be used as an effective predictive tool for the outcome of MVD surgery.


Assuntos
Cirurgia de Descompressão Microvascular , Avaliação de Resultados em Cuidados de Saúde , Neuralgia do Trigêmeo/fisiopatologia , Neuralgia do Trigêmeo/cirurgia , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
7.
J Synchrotron Radiat ; 28(Pt 5): 1662-1668, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34475313

RESUMO

The new Brain Imaging Beamline (BIB) of the Taiwan Photon Source (TPS) has been commissioned and opened to users. The BIB and in particular its endstation are designed to take advantage of bright unmonochromatized synchrotron X-rays and target fast 3D imaging, ∼1 ms exposure time plus very high ∼0.3 µm spatial resolution. A critical step in achieving the planned performances was the solution to the X-ray induced damaging problems of the detection system. High-energy photons were identified as their principal cause and were solved by combining tailored filters/attenuators and a high-energy cut-off mirror. This enabled the tomography acquisition throughput to reach >1 mm3 min-1, a critical performance for large-animal brain mapping and a vital mission of the beamline.


Assuntos
Encéfalo/diagnóstico por imagem , Imageamento Tridimensional , Lesões por Radiação/prevenção & controle , Microtomografia por Raio-X/instrumentação , Animais , Desenho de Equipamento , Fótons , Síncrotrons , Taiwan
8.
J Chin Med Assoc ; 84(11): 1019-1022, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34261980

RESUMO

BACKGROUND: Dopamine agonists such as bromocriptine and cabergoline have been found to be an effective treatment for hyperprolactinemia, not only inducing adenoma shrinkage but also lowering serum prolactin levels. Among known dopamine agonists, cabergoline is the drug of choice due to its enhanced tolerability compared with bromocriptine. This study aimed to evaluate cabergoline's effectiveness, along with transsphenoidal surgery, in the treatment of hyperprolactinemia. METHODS: We retrieved all patients with a diagnosis of prolactinoma who were treated in our hospital during 2000-2018. A total of 208 patients were enrolled in the analysis after applying exclusion criteria. Patients were divided into four groups according to the treatments received. The demographic data, dosage and duration of cabergoline, and serum prolactin levels at different time points were collected for analysis. RESULTS: Normalization was achieved in 59 patients (83.10%) within a short median duration of 2.80 months among those treated with cabergoline only. Although cabergoline alone was effective and well-tolerated, our data showed that long-term remission rates were more favorable when surgery was involved. The long-term remission rate of all patients enrolled was 53.8% (112 patients among 208 patients). The long-term remission rates for the different treatment groups were 17.8% (8 of 45 patients) in Group 1 (Operation→Drug), 83.3% (5 of 6 patients) in Group 2 (Drug→Operation), 79.0% (68 of 86 patients) in Group 3 (Operation only), and 43.7% (31 of 71 patients) in Group 4 (Drug only). CONCLUSION: Cabergoline has been demonstrated to be effective and should be considered as a first-line treatment for hyperprolactinemia. In our study, transsphenoidal surgery was also demonstrated to achieve good results compared with medical treatment. Surgical intervention may resurface as an alternative first-line treatment. When used in combination with cabergoline, surgery offers a higher disease remission rate than either drug or operation alone.


Assuntos
Cabergolina/uso terapêutico , Agonistas de Dopamina/uso terapêutico , Hiperprolactinemia/tratamento farmacológico , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
9.
Biomater Sci ; 9(11): 3979-3988, 2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34085077

RESUMO

The radio-resistance of tumor tissues has been considered a great challenge for cancer radiotherapy (RT).The development of nanoparticle (NP)-based radio-sensitizers can enhance the radio-sensitization of tumor tissues while reducing the side effects to surrounding tissues. However, most of the nano-radiosensitizers show increased radiation deposition with a high-Z element but achieve limited enhancement. Herein, we investigated polyethylene glycol (PEG)-modified gold-iron selenide nanocomposites (Au-FeSe2 NCs) for simultaneously enhancing therapeutic effects in multiple ways. In this study, the high-Z element Au (Z = 79) endows Au-FeSe2 NCs with enhanced X-ray deposition and thus causes more DNA damage. On the other hand, Au-FeSe2 exhibits the ability to produce reactive oxygen species (ROS) by catalyzing endogenous hydrogen peroxide in tumor sites as well as improve the hydrogen peroxide level during ionizing irradiation. Finally, combined with photothermal therapy (PTT), Au-FeSe2 NCs could exhibit a remarkable RT/PTT synergistic effect on tumor treatment.


Assuntos
Nanopartículas Metálicas , Nanocompostos , Linhagem Celular Tumoral , Ouro , Ferro , Estresse Oxidativo
10.
Adv Healthc Mater ; 9(20): e2000864, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32945131

RESUMO

Ferroptotic cell death results from glutathione peroxidase 4 (GPX4) inactivation and/or glutathione (GSH) depletion. Elevated GSH levels are often found in multidrug-resistant (MDR) tumor cells, reducing their sensitivity to chemotherapeutic drugs and the efficacy of treatment. MDR cells also acquire a dependency on GPX4, reducing their oxidative stress and promoting their survival. Therefore, the depletion of GSH and inactivation of GPX4 has the potential to be a superior treatment strategy for MDR tumors. Platinum-decorated gold nanostars (Pt-AuNS) are presented as a novel metal nanoprodrug for ferroptotic therapy against MDR tumors. Under dark conditions, the synthesized Pt-AuNS exhibit negligible levels of toxicity. Upon exposure of the Pt-AuNS to near-infrared (NIR) light, active metallic (Pt and Au) species are released, subsequently inducing cytotoxicity. The mechanism of action is attributed to GSH depletion and GPX4 inactivation, accumulating lipid hydroperoxides, which in turn leads to ferroptosis. In in vivo xenograft, the MDR cancer model confirmed the NIR light-activation of Pt-AuNS prodrugs, resulting in efficient ferroptotic therapeutic action against MDR tumors without long-term side effects. The findings lay the groundwork for using Pt-AuNS prodrugs responsive to NIR light as ferroptosis-inducing agents in chemo-resistant cancer cells and demonstrate their potential for use in future clinical applications.


Assuntos
Ferroptose , Neoplasias , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Ouro , Platina
11.
ACS Appl Mater Interfaces ; 12(27): 30021-30030, 2020 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-32594734

RESUMO

Combining photothermal and photodynamic modalities has shown encouraging therapeutic efficacy against various malignant cancers. Developing a delivery method for targeting and penetrating tumors is still a major focus for advancing this therapeutic approach. Herein, we report a novel strategy involving the utilization of stem cells as a live carrier to codeliver photothermal and photodynamic agents for cancer therapy. To this end, a novel gold nanorod (AuNR)-PEG-PEI (APP)/chlorin e6 (Ce6)-loaded adipose-derived stem cell (ADSC) system is proposed in which AuNRs and Ce6 act as the photothermal and photodynamic agents, respectively. To integrate with stem cells, the APP/Ce6 nanocomplexes exhibit advantages of low drug leakage, low cytotoxicity, efficient cellular uptake, and redox-responsive release. After loading of APP/Ce6 nanocomplexes, the ADSCs still maintained good tumor tropism and were capable of penetrating into the tumor spheroids. The photothermal effect induced by exposure to near-infrared light irradiation at 808 nm promoted the release of Ce6 from the stem cells into the surroundings and hence increased its availability to treat cancer cells. APP/Ce6-loaded ADSCs exerted effective dose-dependent in vitro anticancer activities via anticipated photothermal and photodynamic effects. In a murine CT26 colon cancer model, APP/Ce6 delivered by ADSCs resulted in superior tumor suppression compared to other delivery strategies. It was also noted that in vivo applications of APP/Ce6-loaded ADSCs did not induce noticeable detrimental effects on normal tissues/organs.


Assuntos
Ouro/química , Fotoquimioterapia/métodos , Porfirinas/química , Células-Tronco/citologia , Células-Tronco/metabolismo , Tecido Adiposo/citologia , Animais , Linhagem Celular Tumoral , Clorofilídeos , Camundongos
12.
Biomater Sci ; 8(7): 1934-1950, 2020 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32039412

RESUMO

An aptamer-conjugated gold nanostar (dsDDA-AuNS) has been developed for targeting nucleolin present in both tumor cells and tumor vasculature for conducting a drug-resistant cancer therapy. AuNS with its strong absorption in the near-infrared (NIR) region was assembled with a layer of the anti-nucleolin aptamer AS1411. An anticancer drug, namely doxorubicin (DOX), was specifically conjugated on deoxyguanosine residues employing heat and acid labile methylene linkages. In response to NIR irradiation, dsDDA-AuNS allowed on-demand therapeutics. AS1411 played an active role in drug cargo-nucleus interactions, enhancing drug accumulation in the nuclei of drug-resistant breast cancer cells. The intravenous injection of dsDDA-AuNS allowed higher drug accumulation in drug-resistant tumors over naked drugs, leading to greater therapeutic efficacy even at a 54-fold less equivalent drug dose. The in vivo triggered release of DOX from dsDDA-AuNS was achieved by NIR irradiation, resulting in simultaneous photothermal and chemotherapeutic actions, yielding superior tumor growth inhibition than those obtained from either type of monotherapy for overcoming drug resistance in cancers.


Assuntos
Antineoplásicos/administração & dosagem , Aptâmeros de Nucleotídeos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ouro/química , Oligodesoxirribonucleotídeos/administração & dosagem , Administração Intravenosa , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Feminino , Humanos , Células MCF-7 , Nanopartículas Metálicas , Camundongos , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/farmacologia , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/genética , Proteínas de Ligação a RNA/efeitos dos fármacos , Proteínas de Ligação a RNA/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Nucleolina
13.
Nanotheranostics ; 3(3): 266-283, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31263658

RESUMO

Cancer combination therapy can improve treatment efficacy and is widely utilized in the biomedical field. In this paper, we propose a facile strategy to develop a polydopamine (PDA)-coated Au nanostar (NS@PPFA) as a multifunctional nanoplatform for cancer targeting and combination therapy. The Au nanostar demonstrated high photothermal conversion efficiency because of the tip-enhanced plasmonic effect. Modification of PDA and folic acid on the NS surface improved its drug-loading efficiency and targeting capability. In vitro, compared with nontargeted cells, targeted breast cancer MCF-7 cells demonstrated efficient uptake of chemodrug-loaded NS-D@PPFA through the receptor-mediated endocytosis pathway. In combination with the photothermal effect induced by near-infrared laser irradiation, controlled payload release could be activated in response to both internal (acid) and external (photothermal) stimuli, leading to an efficient chemo-photothermal action against MCF-7 cells and drug-resistant MCF-7/ADR cells. By contrast, cellular damage was less obvious in normal HaCaT (human skin keratinocytes) and NIH-3T3 cells (murine fibroblasts). In addition, payload-free NS@PPFA exhibited a high binding affinity (Kd = 2.68 × 10-10 M) toward vascular endothelial growth factor (VEGF-A165), which was at least two orders of magnitude stronger than that of highly abundant plasma proteins, such as human serum albumin. Furthermore, in vitro study showed that NS@PPFA could effectively inhibit VEGF-A165-induced proliferation, migration, and tube formation of human umbilical vein endothelial cells, resulting in additional therapeutic benefits for eradicating tumors through a simultaneous antiangiogenic action in chemo-photothermal treatment. The combined treatment also exhibited the lowest microvessel density, leading to a potent antitumor effect in vivo. Overall, our "all-in-one" nanoplatform is highly promising for tumor therapy, enabling effective treatment against multidrug-resistant cancers.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Ouro/química , Indóis/química , Nanopartículas/química , Polímeros/química , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Liberação Controlada de Fármacos , Endocitose/efeitos dos fármacos , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Hipertermia Induzida , Células MCF-7 , Camundongos , Camundongos Nus , Células NIH 3T3 , Nanopartículas/ultraestrutura , Neovascularização Fisiológica/efeitos dos fármacos , Fototerapia , Temperatura , Fatores de Tempo , Distribuição Tecidual/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Nanoscale ; 11(12): 5580-5594, 2019 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-30860532

RESUMO

We have developed a rapid and straightforward topical treatment method for dry eye disease (DED) using poly(catechin) capped-gold nanoparticles (Au@Poly-CH NPs) carrying amfenac [AF; a nonsteroidal anti-inflammatory drug (NSAID)] through effective attenuation of ocular surface tissue damage in dry eyes. A dual-targeted strategy based on ocular therapeutics was adopted to simultaneously block the cyclooxygenase enzymes-induced inflammation and reactive oxygen species (ROS)-induced oxidative stress, the primary two causes of DED. The self-assembled core-shell Au@Poly-CH NPs synthesized via a simple reaction between tetrachloroaurate(iii) and catechin possess a poly(catechin) shell (∼20 nm) on the surface of each Au NP (∼60 nm). The anti-oxidant and anti-inflammatory properties of AF/Au@Poly-CH NPs were evaluated by DCFH-DA and prostaglandin E2/VEGF assays, respectively. Our results demonstrate that Au@Poly-CH NPs not only act as an anti-oxidant to suppress ROS-mediated processes, but also serve as a drug carrier of AF for a synergistic effect on anti-inflammation. In vivo biocompatibility studies show good tolerability of AF/Au@Poly-CH NPs for potential use in the treatment of ocular surface pathologies. The dual-targeted therapeutic effects of AF/Au@Poly-CH NPs lead to rapid recovery from DED in a rabbit model. Au@Poly-CH NPs loaded with NSAIDs is a promising multifunctional nanocomposite for treating various inflammation- and oxidative stress-related diseases.


Assuntos
Anti-Inflamatórios/química , Antioxidantes/química , Nanopartículas Metálicas/química , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Materiais Biocompatíveis/química , Catequina/química , Linhagem Celular , Córnea/citologia , Córnea/metabolismo , Córnea/patologia , Liberação Controlada de Fármacos , Síndromes do Olho Seco/tratamento farmacológico , Síndromes do Olho Seco/patologia , Ouro/química , Microscopia de Fluorescência , Mucina-5AC/metabolismo , Soluções Oftálmicas/química , Soluções Oftálmicas/farmacologia , Soluções Oftálmicas/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Fenilacetatos/química , Fenilacetatos/farmacologia , Fenilacetatos/uso terapêutico , Prostaglandina-Endoperóxido Sintases/química , Prostaglandina-Endoperóxido Sintases/metabolismo , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
J Microbiol Immunol Infect ; 51(6): 829-838, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30196007

RESUMO

BACKGROUND/PURPOSE: Dengue disease is widespread in tropical and sub-tropical regions. Severe dengue infection is characterized by plasma leakage, fluid accumulation, severe bleeding, or vital organ impairment. Bleeding is a critical complication of dengue disease. However, the biomarkers of dengue disease are still unknown. Macrophages have a distinct polarization phenotype related to M1/M2 classification. Macrophage polarization toward the pro-inflammatory M1 phenotype is considered critical for efficient antiviral immune responses, whereas the anti-inflammatory M2 phenotype is considered essential for tissue remodeling. We investigated macrophage polarization patterns in the peripheral blood of pediatric patients with dengue disease. METHODS: Medical records and laboratory data were collected from 23 pediatric healthy controls and 100 dengue disease samples from 50 dengue patients. Macrophage polarization-related surface markers were assessed using flow cytometry. RESULTS: The percentage of macrophages in the peripheral blood was higher in dengue patients than in the healthy controls. The percentages of M2a and M2c macrophage subsets were higher and the percentage of M1 macrophage subset was lower in dengue patients than in healthy controls. However, the percentages of M1, M2a and M2b macrophage subsets in dengue patients with bleeding tendency were lower than that without bleeding tendency. The percentages of M2a, M2b, and M2c macrophage subsets were positively correlated with platelet counts. CONCLUSION: Decreased the percentages of M2 macrophage subsets in pediatric dengue patients are associated with bleeding tendency and lower platelet counts.


Assuntos
Dengue/sangue , Dengue/complicações , Hemorragia/sangue , Hemorragia/etiologia , Macrófagos/imunologia , Adolescente , Adulto , Biomarcadores/metabolismo , Contagem de Células Sanguíneas , Criança , Pré-Escolar , Dengue/patologia , Feminino , Humanos , Lactente , Ativação de Macrófagos , Macrófagos/citologia , Masculino , Fenótipo , Contagem de Plaquetas , Taiwan , Adulto Jovem
16.
ACS Appl Mater Interfaces ; 10(25): 21160-21172, 2018 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-29863836

RESUMO

Abnormal biochemical alteration such as unbalanced reactive oxygen species (ROS) levels has been considered as a potential disease-specific trigger to deliver therapeutics to target sites. However, in view of their minute variations in concentration, short lifetimes, and limited ranges of action, in situ generation of ROS with specific manipulations should be more effective for ROS-responsive drug delivery. Here we present a new delivery nanoplatform for photodynamic therapy (PDT) with on-demand drug release regulated by light irradiation. Rose bengal (RB) molecules, which exhibit a high yield of ROS generation, were encapsulated in a mixture of chitosan (CTS), poly(vinyl alcohol) (PVA), and branched polyethylenimine ( bPEI) with hydrophobic iron oxide nanoparticles through an oil-in-water emulsion method. The as-prepared magnetic nanoclusters (MNCs) with a tripolymer coating displayed high water dispersibility, efficient cellular uptake, and the cationic groups of CTS and bPEI were effective for RB loading through electrostatic interaction. The encapsulation efficiency of RB in MNCs could be further improved by increasing the amount of short bPEI chains. During the photodynamic process, controlled release of the host molecules (i.e., RB) or guest molecules (i.e., paclitaxel) from the bPEI-based nanoplatform was achieved simultaneously through a photooxidation action sensitized by RB. This approach promises specific payload release and highly effective PDT or PDT combined therapy in various cancer cell lines including breast (MCF-7 and multidrug resistant MCF-7 subline), SKOV-3 ovarian, and Tramp-C1 prostate. In in vivo xenograft studies, the nanoengineered light-switchable carrier also greatly augments its PDT efficacy against multidrug resistant MCF-7/MDR tumor as compared with free drugs. All the above findings suggest that the substantial effects of enhanced drug distribution for efficient cancer therapy was achieved with this smart nanocarrier capable of on demand drug release and delivery, thus exerting its therapeutic activity to a greater extent.


Assuntos
Liberação Controlada de Fármacos , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Masculino , Nanopartículas , Fotoquimioterapia , Rosa Bengala
17.
Int J Colorectal Dis ; 33(7): 985-989, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29619561

RESUMO

PURPOSE: The role of adjuvant chemotherapy (ACT) in treating patients who have colorectal liver metastases (CLM) and undergo liver metastasectomy (LMS) is unclear in this patient population. We aimed to compare the mortality of patients receiving different ACT (i.e., oxaliplatin-based, irinotecan-based, and 5-fluorouracil-only (5FU)) and different treatment frequencies. METHODS: We included 2583 patients with CLM who underwent LMS (including synchronous LMS [SLMS] and metachronous LMS [MLMS]) in this retrospective cohort study. We used Cox proportional hazard model to obtain hazard ratios (HRs) for mortality. The reference group was 5FU-only ACT when comparing ACT type and the reference group was treatment for ≤ 3 times when comparing ACT frequency. RESULTS: In SLMS patients, oxaliplatin-based ACT (HR = 0.78) and receiving ACT for ≥ 4 times (4-6 times, HR = 0.61; 7-9 times, HR = 0.69; 10-12 times, HR = 0.66) were associated with lower risk of mortality. In MLMS patients, oxaliplatin-based ACT (HR = 0.52), irinotecan-based ACT (HR = 0.64), and receiving ACT for 10-12 times (HR = 0.65) were associated with lower risk of mortality. CONCLUSIONS: In SLMS and MLMS patients, patients who received oxaliplatin-based ACT were more likely to survive than patients who received 5FU-only ACT. In MLMS patients, patients who received irinotecan-based ACT were also more likely to survive than those who received 5FU-only ACT. We recommend a course of at least four to six times of ACT after LMS in this patient population.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Hepáticas/secundário , Metastasectomia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimioterapia Adjuvante , Humanos , Neoplasias Hepáticas/cirurgia , Pessoa de Meia-Idade , Compostos Organoplatínicos , Estudos Retrospectivos , Resultado do Tratamento
18.
Front Chem ; 6: 647, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687694

RESUMO

Dual functional drug carrier has been a modern strategy in cancer therapy because it is a platform to elicit additive and synergistic effects through combination therapy. Photo-activated external stimuli such as reactive oxygen species (ROS) also ensure adequate drug delivery in a precise temporal and spatial manner. However, current ROS-responsive drug delivery systems usually require tedious synthetic procedures. A facile one-pot approach has been reported herein, to obtain self-assembled polymeric nanocarriers (NCs) for simultaneous paclitaxel (PTX)- and Rose Bengal (RB)-loading to achieve combined chemo-photodynamic therapy and controlled drug release in responsive to a light-induced ROS stimulus. To encapsulate these hydrophobic and hydrophilic drugs, chitosan (CTS), branched polyethylenimine (bPEI) and polyvinyl alcohol (PVA) were selected and fabricated into nanoblended matrices through an oil-in-water emulsion method. The amphiphilic properties of CTS permit simultaneous entrapment of PTX and RB, while the encapsulation efficiency of RB was further improved by increasing the amount of short-chain bPEI. During the one-step assembly process, bovine serum albumin (BSA) was also added to condense the cationic tripolymer mixtures into more stable nanocarriers (BNCs). Hyaluronic acid (HA) was subsequently grafted onto the surface of BNCs through electrostatic interaction, leading to the formation of HA-BSA/CTS/PVA/bPEI-blended nanocarriers (HBNCs) to achieve an efficient prostate-cancer-cell uptake. Importantly, in response to external light irradiation, HBNCs become destabilized owing to the RB-mediated photodynamic action. It allows an on-demand dual-payload release to evoke a simultaneous photodynamic and chemo treatment for cancer cell eradication. Thus, HBNCs present a new promising approach that exhibits a specific vulnerability to RB-induced photosensitization. The consequent dual-cargo release is also expected to successfully combat cancer through a synergistic anti-tumor effect.

19.
Theranostics ; 7(12): 3034-3052, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28839462

RESUMO

Recently, living cells with tumor-homing properties have provided an exciting opportunity to achieve optimal delivery of nanotherapeutic agents. However, premature payload leakage may impair the host cells, often leading to inadequate in vivo investigations or therapeutic efficacy. Therefore, a nanoplatform that provides a high drug-loading capacity and the precise control of drug release is required. In the present study, a robust one-step synthesis of a doxorubicin (DOX)-loaded gold nanorod/albumin core-shell nanoplatform (NR@DOX:SA) was designed for effective macrophage-mediated delivery to demonstrate how nanoparticle-loaded macrophages improve photothermal/chemodrug distribution and retention ability to achieve enhanced antitumor effects. The serum albumin shell of these nanoagents served as a drug reservoir to delay the intracellular DOX release and drug-related toxicity that impairs the host cell carriers. Near-infrared laser irradiation enabled on-demand payload release to destroy neighboring tumor cells. A series of in vivo quantitative analyses demonstrated that the nanoengineered macrophages delivered the nanodrugs through tumor-tropic migration to tumor tissues, resulting in the twice homogenous and efficient photothermal activations of drug release to treat prostate cancer. By contrast, localized pristine NR@DOX:SAs exhibit limited photothermal drug delivery that further reduces their retention ability and therapeutic efficacy after second combinational treatment, leading to a failure of cancer therapy. Moreover, the resultant unhealable wounds impair quality of life. Free DOX has rapid clearance and therefore exhibits limited antitumor effects. Our findings suggest that in comparison with pristine nanoparticles or free DOX, the nanoengineered macrophages effectively demonstrate the importance and effect of homogeneous drug distribution and retention ability in cancer therapy.


Assuntos
Albuminas/metabolismo , Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Portadores de Fármacos/metabolismo , Ouro/metabolismo , Macrófagos/metabolismo , Nanotubos , Animais , Modelos Animais de Doenças , Tratamento Farmacológico/métodos , Hipertermia Induzida/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/terapia , Células RAW 264.7
20.
Sci Rep ; 7: 41415, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28145512

RESUMO

Chronic pain, resulting from injury, arthritis, and cancer, is often accompanied by inflammation. High concentrations of protons found in inflamed tissues results in tissue acidosis, a major cause of pain and hyperalgesia. Acidosis signals may mediate a transition from acute to chronic hyperalgesia (hyperalgesic priming) via proton-sensing G-protein-coupled receptors (GPCRs). The expression of T-cell death-associated gene 8 (TDAG8), a proton-sensing GPCR, is increased during inflammatory hyperalgesia. Attenuating TDAG8 expression in the spinal cord inhibits bone cancer pain, but whether TDAG8 is involved in inflammatory hyperalgesia or hyperalgesic priming remains unclear. In this study, we used TDAG8-knockout or -knockdown to explore the role of TDAG8 in pain. Suppressed TDAG8 expression delayed the onset of inflammatory hyperalgesia and shortened hyperalgesic time in mice. In a dual acid-injection model (acid [pH 5.0] injected twice, 5 days apart), shRNA inhibition of TDAG8 shortened the duration of the second hyperalgesia. Similar results were found in TDAG8-deficient mice. The dual administration of TDAG8 agonist also confirmed that TDAG8 is involved in hyperalgsic priming. Accordingly, TDAG8 may mediate acidosis signals to initiate inflammatory hyperalgesia and establish hyperalgesic priming.


Assuntos
Hiperalgesia/metabolismo , Hiperalgesia/patologia , Inflamação/metabolismo , Inflamação/patologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Carragenina , AMP Cíclico/metabolismo , Adjuvante de Freund , Deleção de Genes , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Espaço Intracelular/metabolismo , Camundongos Endogâmicos ICR , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA