Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Small ; 19(2): e2205354, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36399643

RESUMO

Durable glioblastoma multiforme (GBM) management requires long-term chemotherapy after surgery to eliminate remaining cancerous tissues. Among chemotherapeutics, temozolomide is considered as the first-line drug for GBM therapy, but the treatment outcome is not satisfactory. Notably, regorafenib, an oral multi-kinase inhibitor, has been reported to exert a markedly superior effect on GBM suppression compared with temozolomide. However, poor site-specific delivery and bioavailability significantly restrict the efficient permeability of regorafenib to brain lesions and compromise its treatment efficacy. Therefore, human H-ferritin (HFn), regorafenib, and Cu2+ are rationally designed as a brain-targeted nanoplatform (HFn-Cu-REGO NPs), fulfilling the task of site-specific delivery and manipulating autophagy and cuproptosis against GBM. Herein, HFn affords a preferential accumulation capacity to GBM due to transferrin receptor 1 (TfR1)-mediated active targeting and pH-responsive delivery behavior. Moreover, regorafenib can inhibit autophagosome-lysosome fusion, resulting in lethal autophagy arrest in GBM cells. Furthermore, Cu2+ not only facilitates the encapsulation of regorafenib to HFn through coordination interaction but also disturbs copper homeostasis for triggering cuproptosis, resulting in a synergistical effect with regorafenib-mediated lethal autophagy arrest against GBM. Therefore, this work may broaden the clinical application scope of Cu2+ and regorafenib in GBM treatment via modulating autophagy and cuproptosis.


Assuntos
Apoptose , Neoplasias Encefálicas , Glioblastoma , Humanos , Apoferritinas , Autofagia , Encéfalo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Cobre
2.
Small ; 19(46): e2303073, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37460404

RESUMO

Glioblastoma (GBM), the most aggressive and lethal form of malignant brain tumor, is a therapeutic challenge due to the drug filtration capabilities of the blood-brain barrier (BBB). Interestingly, glioblastoma tends to resist apoptosis during chemotherapy, but is susceptible to ferroptosis. Developing therapies that can effectively target glioblastoma by crossing the BBB and evoke ferroptosis are, therefore, crucial for improving treatment outcomes. Herein, a versatile biomimetic nanoplatform, L-D-I/NPs, is designed that self-assembled by loading the antimalarial drug dihydroartemisinin (DHA) and the photosensitizer indocyanine green (ICG) onto lactoferrin (LF). This nanoplatform can selectively target glioblastoma by binding to low-density lipoprotein receptor-related protein-1 (LRP1) and crossing the BBB, thus inducing glioblastoma cell ferroptosis by boosting intracellular reactive oxygen species (ROS) accumulation and iron overload. In addition, L-D-I/NPs have demonstrated the ability to effectively suppress the progression of orthotopic glioblastoma and significantly prolong survival in a mouse glioblastoma model. This nanoplatform has facilitated the application of non-chemotherapeutic drugs in tumor treatment with minimal adverse effects, paving the way for highly efficient ferroptosis-based therapies for glioblastoma.


Assuntos
Neoplasias Encefálicas , Ferroptose , Glioblastoma , Glioma , Camundongos , Animais , Glioblastoma/patologia , Reposicionamento de Medicamentos , Barreira Hematoencefálica/metabolismo , Glioma/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral
3.
Int J Mol Sci ; 23(18)2022 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-36142797

RESUMO

Strigolactones (SLs) are a class of important hormones in the regulation of plant branching. In the model plant Arabidopsis, AtMAX1 encodes a cytochrome P450 protein and is a crucial gene in the strigolactone synthesis pathway. Yet, the regulatory mechanism of MAX1 in the shoot branching of wintersweet (Chimonanthus praecox) remains unclear. Here we identified and isolated three MAX1 homologous genes, namely CpMAX1a, CpMAX1b, and CpMAX1c. Quantitative real-time PCR (qRT-PCR) revealed the expression of CpMAX1a in all tissues, being highest in leaves, whereas CpMAX1b was only expressed in stems, while CpMAX1c was expressed in both roots and stem tips. However, CpMAX1a's expression decreased significantly after decapitation; hence, we verified its gene function. CpMAX1a was located in Arabidopsis chloroplasts. Overexpressing CpMAX1a restored the phenotype of the branching mutant max1−3, and reduced the rosette branch number, but resulted in no significant phenotypic differences from the wild type. Additionally, expression of AtBRC1 was significantly upregulated in transgenic lines, indicating that the CpMAX1a gene has a function similar to the homologous gene of Arabidopsis. In conclusion, our study shows that CpMAX1a plays a conserved role in regulating the branch development of wintersweet. This work provides a molecular and theoretical basis for better understanding the branch development of wintersweet.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Calycanthaceae , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Regulação da Expressão Gênica de Plantas , Hormônios/metabolismo , Lactonas/metabolismo , Brotos de Planta/metabolismo
4.
Small ; 16(21): e1907643, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32187839

RESUMO

The safety of metal oxide (MOx) nanoparticles (NPs) has been highly concerned because of their wide application and potential toxicological injury. The safe-by-design strategy is usually developed to make safer MOx NPs based on regulation of their physicochemical properties. In the present study, manganese oxide (Mn3 O4 ) NPs, as a representative of insoluble toxic MOx NPs, are doped with a series of transition metal to regulate their conduction band energy (Ec ) out of biological redox potential range (BRPR) or Fermi energy (Ef ) far away from valence band energy (Ev ), aiming at completely eliminating the toxicity or significantly reducing the toxicity. It is found that all these M-doping cannot move Ec of Mn3 O4 NPs out of the BRPR but zinc (Zn)-, copper (Cu)-, and chromium (Cr)-doping do move Ef far away from Ev , where Zn-doping results in the largest |Ef - Ev | value. Various abiotic, in vitro and in vivo assessments reveal that Zn-, Cu-, and Cr-doped Mn3 O4 NPs can generate lower amount of •OH and trigger weaker injury than Mn3 O4 NPs, where Zn-doped Mn3 O4 NPs show the lowest toxicity. Regulating Ef far away from Ev becomes a feasible safe-by-design approach to achieve safe MOx NPs.

5.
Nano Lett ; 19(7): 4478-4489, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31244230

RESUMO

Silver nanoparticles (Ag NPs) have promising plasmonic properties, however, they are rarely used in biomedical applications because of their potent toxicity. Herein, an electron compensation effect from Au to Ag was applied to design safe Au@Ag core-shell NPs. The Ag shell thickness was precisely regulated to enable the most efficient electron enrichment in Ag shell of Au@Ag2.4 NPs, preventing Ag oxidation and subsequent Ag+ ion release. X-ray photoelectron spectroscopy and X-ray absorption near-edge structure analysis revealed the electron transfer process from Au core to Ag shell, and inductively coupled plasma optical emission spectroscopy analysis confirmed the low Ag+ ion release from Au@Ag2.4 NPs. Bare Au@Ag2.4 NPs showed much lower toxicological responses than Ag NPs in BEAS-2B and Raw 264.7 cells and acute lung inflammation mouse models, and PEGylation of Au@Ag2.4 NPs could further improve their safety to L02 and HEK293T cells as well as mice through intravenous injection. Further, diethylthiatri carbocyanine iodide attached pAu@Ag2.4 NPs exhibited intense surface-enhanced Raman scattering signals and were used for Raman imaging of MCF7 cells and Raman biosensing in MCF7 tumor-bearing mice. This electron compensation effect opens up new opportunity for broadening biomedical application of Ag-based NPs.

6.
Nano Lett ; 19(10): 6800-6811, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31466437

RESUMO

Nitric oxide (NO) molecular messenger can reverse the multidrug resistance (MDR) effect of cancer cells through reducing P-glycoprotein (P-gp) expression, beneficial for creating a favorable microenvironment for the treatment of doxorubicin (Dox)-resistant cancer cells. Development of sophisticated nanosystems to programmably release NO and Dox becomes an efficient strategy to overcome the MDR obstacles and achieve promising therapeutic effects in Dox-resistant cancer. Herein, a NO stimulated nanosystem was designed to engineer a significant time gap between NO and Dox release, promoting MDR cancer therapy. A o-phenylenediamine-containing lipid that can hydrolyze in response to NO was embedded in the phospholipid bilayer structure of liposome to form NO-responsive liposome, which could further encapsulate l-arginine (l-Arg)/Dox-loaded gold@copper sulfide yolk-shell nanoparticls (ADAu@CuS YSNPs) to form ADLAu@CuS YSNPs. Under 808 nm laser irradiation, the unique resonant energy transfer (RET) process and reactive oxygen species (ROS) generation in the confined space of ADLAu@CuS YSNPs could effectively convert l-Arg into NO, regionally destabilizing the phospholipid bilayer structure, as a result of NO release. However, at this early stage Dox could not be released from YSNPs due to the molecular scaffold limit. As the NO release progressed, the NO-responsive liposome layer was deteriorated more severely, allowing Dox to escape. This NO and Dox sequential release of ADLAu@CuS YSNPs could significantly inhibit P-gp expression and enhance Dox accumulation in Dox-resistant MCF-7/ADR cells, leading to promising in vitro and in vivo therapeutic effects and presenting their great potential for MDR cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Preparações de Ação Retardada/metabolismo , Doxorrubicina/administração & dosagem , Óxido Nítrico/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Lipossomos/metabolismo , Células MCF-7 , Nanopartículas/metabolismo
7.
Nano Lett ; 19(6): 4134-4145, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31084016

RESUMO

As a noninvasive treatment modality, ultrasound (US)-triggered sonodynamic therapy (SDT) shows broad and promising applications to overcome the drawbacks of traditional photodynamic therapy (PDT) in combating cancer. However, the SDT efficacy is still not satisfactory without oxygen (O2) assistance. In addition, there is also much space to explore the SDT-based synergistic therapeutic modalities. Herein, a novel Pt-CuS Janus composed of hollow semiconductor CuS and noble metallic Pt was rationally designed and successfully synthesized. The hollow CuS shows a large inner cavity for loading sonosensitizer molecules (tetra-(4-aminophenyl) porphyrin, TAPP) to implement SDT. Moreover, the deposition of Pt not only enhances photothermal performance compared with those of CuS nanoparticles (NPs) due to the effect of the local electric field enhancement but also possesses nanozyme activity for catalyzing decomposition of endogenous overexpressed hydrogen peroxide (H2O2) to produce O2 that can overcome tumor hypoxia and augment the SDT-induced highly toxic reactive oxygen species (ROS) production for efficient cancer cell apoptosis. Importantly, the generated heat of Pt-CuS by 808 nm laser irradiation can accelerate the catalytic activity of Pt and elevate the O2 level that further facilitates SDT efficacy. Interestingly, the thermally sensitive copolymer coated around the Janus can act as a smart switch to regulate the catalytic ability of Pt and control TAPP release that has a significant effect on modulating the therapeutic effect. The synergistic catalysis-enhanced SDT efficiency and highly photothermal effect almost realized complete tumor resection without obvious reoccurrence and simultaneously displayed a highly therapeutic biosafety. Furthermore, the high optical absorbance allows the as-synthesized Pt-CuS Janus for photoacoustic (PA) imaging and NIR thermal imaging. This work develops a versatile nanoplatform for a multifunctional theranostic strategy and broadens the biological applications by rationally designing their structure.


Assuntos
Neoplasias do Colo/terapia , Cobre/uso terapêutico , Nanopartículas/uso terapêutico , Platina/uso terapêutico , Animais , Linhagem Celular Tumoral , Humanos , Hipertermia Induzida , Camundongos , Nanopartículas/ultraestrutura , Hipóxia Tumoral , Terapia por Ultrassom
8.
Expert Rev Proteomics ; 16(5): 413-429, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30925852

RESUMO

INTRODUCTION: Resistance to chemotherapy and development of specific and effective molecular targeted therapies are major obstacles facing current cancer treatment. Comparative proteomic approaches have been employed for the discovery of putative biomarkers associated with cancer drug resistance and have yielded a number of candidate proteins, showing great promise for both novel drug target identification and personalized medicine for the treatment of drug-resistant cancer. Areas covered: Herein, we review the recent advances and challenges in proteomics studies on cancer drug resistance with an emphasis on biomarker discovery, as well as understanding the interconnectivity of proteins in disease-related signaling pathways. In addition, we highlight the critical role that post-translational modifications (PTMs) play in the mechanisms of cancer drug resistance. Expert opinion: Revealing changes in proteome profiles and the role of PTMs in drug-resistant cancer is key to deciphering the mechanisms of treatment resistance. With the development of sensitive and specific mass spectrometry (MS)-based proteomics and related technologies, it is now possible to investigate in depth potential biomarkers and the molecular mechanisms of cancer drug resistance, assisting the development of individualized therapeutic strategies for cancer patients.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteômica , Animais , Biomarcadores Tumorais/metabolismo , Humanos , Espectrometria de Massas , Processamento de Proteína Pós-Traducional
9.
Nano Lett ; 18(2): 886-897, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29323915

RESUMO

Gold (Au) core@void@copper sulfide (CuS) shell (Au-CuS) yolk-shell nanoparticles (YSNPs) were prepared in the present study for potential chemo-, photothermal, and photodynamic combination therapy, so-called "chemophototherapy". The resonance energy transfer (RET) process was utilized in Au-CuS YSNPs to achieve both enhanced photothermal and photodynamic performance compared with those of CuS hollow nanoparticles (HNPs). A series of Au nanomaterials as cores that had different localized surface plasmon resonance (LSPR) absorption peaks at 520, 700, 808, 860, and 980 nm were embedded in CuS HNPs to screen the most effective Au-CuS YSNPs according to the RET process. Thermoresponsive polymer was fabricated on these YSNPs' surface to allow for controlled drug release. Au808-CuS and Au980-CuS YSNPs were found capable of inducing the largest temperature elevation and producing the most significant hydroxyl radicals under 808 and 980 nm laser irradiation, respectively, which could accordingly cause the most severe 4T1 cell injury through oxidative stress mechanism. Moreover, doxorubicin-loaded (Dox-loaded) P(NIPAM-co-AM)-coated Au980-CuS (p-Au980-CuS@Dox) YSNPs could more efficiently kill cells than unloaded particles upon 980 nm laser irradiation. After intravenous administration to 4T1 tumor-bearing mice, p-Au980-CuS YSNPs could significantly accumulate in the tumor and effectively inhibit the tumor growth after 980 nm laser irradiation, and p-Au980-CuS@Dox YSNPs could further potentiate the inhibition efficiency and exhibit excellent in vivo biocompatibility. Taken together, this study sheds light on the rational design of Au-CuS YSNPs to offer a promising candidate for chemophototherapy.


Assuntos
Cobre/uso terapêutico , Ouro/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/terapia , Sulfetos/uso terapêutico , Animais , Linhagem Celular Tumoral , Cobre/administração & dosagem , Preparações de Ação Retardada/química , Doxorrubicina/administração & dosagem , Doxorrubicina/uso terapêutico , Transferência de Energia , Ouro/administração & dosagem , Hipertermia Induzida/métodos , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Neoplasias/patologia , Fotoquimioterapia/métodos , Sulfetos/administração & dosagem
10.
Chemistry ; 24(66): 17405-17418, 2018 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-29876975

RESUMO

Theranostic nanomaterials (NMs) have gained increasing attention for their simultaneous performance of diagnosis and therapy. Bi-based NMs hold great potential as theranostic platforms based on their X-ray sensitive capability, near-infrared driven semiconductor properties, and distinctive structures, which facilitate the computed tomography (CT) imaging, photoacoustic (PA) imaging, radiation therapy, and phototherapy. The sophisticated design in composition, structure, and surface fabrication of Bi-based NMs can endow these NMs with more modalities in cancer diagnosis and therapy. In this Minireview, we focus on the recent advances in Bi-based theranostic NMs. A series of unique structures and functions as well as the underlying property-activity relationship of Bi-based NMs are showcased to highlight their promising imaging and therapeutic performance. At the end, we propose some challenges for the design and preparation of Bi-based NMs to improve their cancer diagnostic and therapeutic performance.


Assuntos
Bismuto/química , Nanoestruturas/química , Neoplasias/diagnóstico , Animais , Humanos , Neoplasias/radioterapia , Técnicas Fotoacústicas , Fototerapia , Espécies Reativas de Oxigênio/metabolismo , Nanomedicina Teranóstica , Tomografia Computadorizada por Raios X
11.
Ecotoxicol Environ Saf ; 163: 612-619, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30092543

RESUMO

Airborne fine particulate matter (PM2.5) has been known capable of causing lung inflammation and fibrosis, as a result of a series of chronic respiration diseases. Although NLRP3 inflammasome activation is essential for development of many chronic diseases, the relationship between PM2.5-induced toxicological effect and NLRP3 inflammasome activation is rarely investigated. Since PM2.5 contains a large population of nanosized materials and many types of nanomaterials can activate NLRP3 inflammasome, the NLRP3 inflammasome activation and lung fibrosis induced by PM2.5 were investigated in the present study. PM2.5 was found capable of causing weak cell death but potent IL-1ß secretion in THP-1 cells, which was involved in NLRP3 inflammasome activation as evidenced by Z-YVAD-FMK inhibited IL-1ß secretion and overexpressed ASC and NLRP3 protein in PM2.5 treated cells. PM2.5 could be internalized into cells through multiple endocytosis processes, such as phagocytosis and pinocytosis (macropinocytosis, clathrin- and caveolin-mediated endocytosis), and activate NLRP3 inflammasome through cathepsin B release, ROS production, and potassium efflux. After 21 days of exposure to PM2.5 through oropharyngeal aspiration, Balb/c mice showed increased IL-1ß and TGF-ß1 levels in the bronchoalveolar lavage fluid (BALF) of lung and significant collagen deposition around small airways of mice, suggesting potential lung inflammation and pulmonary fibrosis.


Assuntos
Inflamassomos/metabolismo , Pulmão/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Material Particulado/toxicidade , Fibrose Pulmonar/induzido quimicamente , Animais , Linhagem Celular , Humanos , Inflamassomos/fisiologia , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/induzido quimicamente
12.
Angew Chem Int Ed Engl ; 57(1): 246-251, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29139182

RESUMO

Bismuth sulfide (Bi2 S3 ) nanomaterials are emerging as a promising theranostic platform for computed tomography imaging and photothermal therapy of cancer. Herein, the photothermal properties of Bi2 S3 nanorods (NRs) were unveiled to intensely correlate to their intrinsic deep-level defects (DLDs) that potentially could work as electron-hole nonradiative recombination centers to promote phonon production, ultimately leading to photothermal performance. Bi2 S3 -Au heterojunction NRs were designed to hold more significant DLD properties, exhibiting more potent photothermal performance than Bi2 S3 NRs. Under 808 nm laser irradiation, Bi2 S3 -Au NRs could trigger higher cellular heat shock protein 70 expression and more apoptotic cells than Bi2 S3 NRs, and caused severe cell death and tumor growth inhibition, showing great potential for photothermal therapy of cancer guided by computed tomography imaging.


Assuntos
Bismuto/química , Ouro/química , Hipertermia Induzida , Nanotubos , Neoplasias/terapia , Fototerapia , Sulfetos/química , Tomografia Computadorizada por Raios X/métodos , Animais , Linhagem Celular Tumoral , Terapia Combinada , Proteínas de Choque Térmico HSP70/metabolismo , Xenoenxertos , Humanos , Raios Infravermelhos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Espectroscopia Fotoeletrônica , Espectrofotometria Ultravioleta , Nanomedicina Teranóstica/métodos
13.
Small ; 13(20)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28371113

RESUMO

Simulated sunlight has promise as a light source able to alleviate the severe pain associated with patients during photodynamic therapy (PDT); however, low sunlight utilization efficiency of traditional photosensitizers dramatically limits its application. Titanium-dioxide-nanoparticle-gold-nanocluster-graphene (TAG) heterogeneous nanocomposites are designed to efficiently utilize simulated sunlight for melanoma skin cancer PDT. The narrow band gap in gold nanoclusters (Au NCs), and staggered energy bands between Au NCs, titanium dioxide nanoparticles (TiO2 NPs), and graphene can result in efficient utilization of simulated sunlight and separation of electron-hole pairs, facilitating the production of abundant hydroxyl and superoxide radicals. Under irradiation of simulated sunlight, TAG nanocomposites can trigger a series of toxicological responses in mouse B16F1 melanoma cells, such as intracellular reactive oxygen species production, glutathione depletion, heme oxygenase-1 expression, and mitochondrial dysfunctions, resulting in severe cell death. Furthermore, intravenous or intratumoral administration of biocompatible TAG nanocomposites in B16F1-tumor-xenograft-bearing mice can significantly inhibit tumor growth and cause severe pathological tumor tissue changes. All of these results demonstrate prominent simulated sunlight-mediated PDT effects.


Assuntos
Ouro/química , Melanoma/tratamento farmacológico , Nanocompostos/química , Nanopartículas/química , Fotoquimioterapia , Neoplasias Cutâneas/tratamento farmacológico , Luz Solar , Titânio/uso terapêutico , Animais , Feminino , Ouro/administração & dosagem , Grafite/administração & dosagem , Grafite/química , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos Endogâmicos BALB C , Nanocompostos/administração & dosagem , Nanocompostos/ultraestrutura , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Titânio/administração & dosagem
14.
Expert Rev Proteomics ; 14(4): 351-362, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28276747

RESUMO

INTRODUCTION: Chemoresistance is a major obstacle for current cancer treatment. Proteogenomics is a powerful multi-omics research field that uses customized protein sequence databases generated by genomic and transcriptomic information to identify novel genes (e.g. noncoding, mutation and fusion genes) from mass spectrometry-based proteomic data. By identifying aberrations that are differentially expressed between tumor and normal pairs, this approach can also be applied to validate protein variants in cancer, which may reveal the response to drug treatment. Areas covered: In this review, we will present recent advances in proteogenomic investigations of cancer drug resistance with an emphasis on integrative proteogenomic pipelines and the biomarker discovery which contributes to achieving the goal of using precision/personalized medicine for cancer treatment. Expert commentary: The discovery and comprehensive understanding of potential biomarkers help identify the cohort of patients who may benefit from particular treatments, and will assist real-time clinical decision-making to maximize therapeutic efficacy and minimize adverse effects. With the development of MS-based proteomics and NGS-based sequencing, a growing number of proteogenomic tools are being developed specifically to investigate cancer drug resistance.


Assuntos
Biomarcadores Tumorais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias/genética , Proteogenômica , Genoma Humano , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Proteoma/genética , Proteômica
15.
Expert Rev Proteomics ; 13(4): 367-81, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26923776

RESUMO

Cancer is a common disease that is a leading cause of death worldwide. Currently, early detection and novel therapeutic strategies are urgently needed for more effective management of cancer. Importantly, protein profiling using clinical proteomic strategies, with spectacular sensitivity and precision, offer excellent promise for the identification of potential biomarkers that would direct the development of targeted therapeutic anticancer drugs for precision medicine. In particular, clinical sample sources, including tumor tissues and body fluids (blood, feces, urine and saliva), have been widely investigated using modern high-throughput mass spectrometry-based proteomic approaches combined with bioinformatic analysis, to pursue the possibilities of precision medicine for targeted cancer therapy. Discussed in this review are the current advantages and limitations of clinical proteomics, the available strategies of clinical proteomics for the management of precision medicine, as well as the challenges and future perspectives of clinical proteomics-driven precision medicine for targeted cancer therapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias/diagnóstico , Medicina de Precisão/métodos , Proteômica/métodos , Animais , Humanos , Terapia de Alvo Molecular/métodos , Neoplasias/metabolismo , Neoplasias/terapia , Medicina de Precisão/tendências
16.
J Oral Maxillofac Surg ; 74(4): 844-50, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26541225

RESUMO

PURPOSE: MicroRNA-340 (miR-340) is deregulated in many human cancers in correlation with tumor progression. Recent studies have found that microRNAs play key roles in energy metabolism. This study explored the contributions of miR-340 to the metabolic shift in oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS: MiR-340 expression was measured by real-time polymerase chain reaction. MiR-340 mimics, miR-340 inhibitor, and scramble small interfering RNA were transfected into SAS human tongue SCC cells to observe their effects on cell proliferation, colony formation, lactate secretion, and glucose uptake rate. Moreover, the relation between the level of miR-340 and glucose transporter-1 (Glut1) was investigated. RESULTS: The expression of miR-340 was decreased and thus induced a metabolic switch in oral cancer cells. The decrease in miR-340 increased Glut1 expression, leading to an increase in lactate secretion and glucose uptake rate. The altered metabolism induced by miR-340 resulted in the rapid proliferation of oral cancer cells. CONCLUSION: The findings suggest that miR-340 might act as a molecular switch that contributes to the regulation of glycolysis in OSCC by regulating Glut1 expression.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Transportador de Glucose Tipo 1/fisiologia , MicroRNAs/fisiologia , Neoplasias da Língua/metabolismo , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/fisiologia , Células Epiteliais/metabolismo , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Glicólise/fisiologia , Células HEK293 , Humanos , Ácido Láctico/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Mucosa Bucal/citologia , Mucosa Bucal/metabolismo , Plasmídeos/genética , RNA Interferente Pequeno/fisiologia , Transfecção
17.
J Asian Nat Prod Res ; 18(9): 908-12, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27268073

RESUMO

A new diterpenoid alkaloid, named bullatine H (1), along with 10 known diterpenoid alkaloids were isolated from the roots of Aconitum brachypodum Diels (Ranunculaceae). The structure of 1 was elucidated by analysis of its spectroscopic data. It should be noted that compound 1 is the first example with 11, 13-dioxygenated denudatine-type diterpenoid alkaloid isolated from Aconitum brachypodum.


Assuntos
Aconitum/química , Alcaloides/isolamento & purificação , Diterpenos/isolamento & purificação , Medicamentos de Ervas Chinesas/isolamento & purificação , Raízes de Plantas/química , Alcaloides/química , Diterpenos/química , Medicamentos de Ervas Chinesas/química , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular
18.
J Proteome Res ; 14(6): 2480-91, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25928036

RESUMO

Paclitaxel (PTX) is a widely used chemotherapeutic drug effective against numerous cancers. To elucidate cellular pathways targeted by PTX and identify novel mechanisms of PTX resistance, we used a SILAC based quantitative proteomic approach to evaluate global changes of cellular protein abundance in HeLa cells. We identified 347 proteins involved in a number of biological processes including spindle assembly, mitotic exit, and extracellular adhesion whose abundance changes upon PTX treatment. Notably, the tumor suppressor PDCD4 involved in translation suppression was down-regulated by PTX. We demonstrated that PDCD4 is a cell-cycle regulated protein and that changes in its abundance are sufficient to alter PTX sensitivity in multiple human cancer cell lines. Immunoprecipitation of PDCD4-RNA complexes and RT-PCR revealed that PDCD4 mediated PTX sensitivity acts through its interaction with mRNA of UBE2S, a ubiquitin K11 linkage conjugating enzyme critical for mitotic exit. Lastly, high levels of PDCD4 in lung cancer tissues are positively correlated with the longer overall survival time of the examined lung cancer patients with PTX involved adjuvant therapy. Therefore, our proteomic screen for paclitaxel targets not only provided novel insight into the cellular resistance to paclitaxel via the PDCD4-mitotic exit regulation axis, but also offered a predictive biomarker for paclitaxel-based personalized chemotherapy in the treatment of lung cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Proteínas Reguladoras de Apoptose/fisiologia , Paclitaxel/farmacologia , Proteômica , Proteínas de Ligação a RNA/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , China , Sistemas de Gerenciamento de Base de Dados , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Análise de Sobrevida
19.
Small ; 11(31): 3797-805, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25930061

RESUMO

While it is well known that there are interspecies differences in Ag sensitivity, differences in the cytotoxic responses of mammalian cells to silver nanoparticles (Ag NPs) are also observed. In order to explore these response outcomes, six cell lines, including epithelial cells (Caco-2, NHBE, RLE-6TN, and BEAS-2B) and macrophages (RAW 264.7 and THP-1) of human and rodent origin, are exposed to 20 nm citrate- and PVP-coated Ag NPs with Au cores, as well as 20 nm citrate-coated particles without cores. An MTS assay shows that while Caco-2 and NHBE cells are resistant to particles over a 0.1-50 µg mL(-1) dose range, RAW 264.7, THP-1, RLE-6TN, and BEAS-2B cells are more susceptible. While there are small differences in dissolution rates, there are no major differences in the cytotoxic potential of the different particles. However, differences in anti-oxidant defense and metallothionein expression among different cell types are observed, which can partially explain differential Ag NP sensitivity. So, it is important to consider these differences in understanding the potential heterogeneous effects of nano Ag on mammalian biological systems.


Assuntos
Antioxidantes/química , Nanopartículas Metálicas/química , Metalotioneína/química , Prata/química , Animais , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Meios de Cultura/química , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Glutationa Transferase/metabolismo , Ouro/química , Humanos , Hidrodinâmica , Macrófagos/metabolismo , Camundongos , Estresse Oxidativo , Ratos
20.
Small ; 11(17): 2087-97, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25581126

RESUMO

The purpose of this paper is to elucidate the key role of NADPH oxidase in NLRP3 inflammasome activation and generation of pulmonary fibrosis by multi-walled carbon nanotubes (MWCNTs). Although it is known that oxidative stress plays a role in pulmonary fibrosis by single-walled CNTs, the role of specific sources of reactive oxygen species, including NADPH oxidase, in inflammasome activation remains to be clarified. In this study, three long aspect ratio (LAR) materials (MWCNTs, single-walled carbon nanotubes, and silver nanowires) are used to compare with spherical carbon black and silver nanoparticles for their ability to trigger oxygen burst activity and NLRP3 assembly. All LAR materials but not spherical nanoparticles induce robust NADPH oxidase activation and respiratory burst activity in THP-1 cells, which are blunted in p22(phox) -deficient cells. The NADPH oxidase is directly involved in lysosomal damage by LAR materials, as demonstrated by decreased cathepsin B release and IL-1ß production in p22(phox) -deficient cells. Reduced respiratory burst activity and inflammasome activation are also observed in bone marrow-derived macrophages from p47(phox) -deficient mice. Moreover, p47(phox) -deficient mice have reduced IL-1ß production and lung collagen deposition in response to MWCNTs. Lung fibrosis is also suppressed by N-acetyl-cysteine in wild-type animals exposed to MWCNTs.


Assuntos
Proteínas de Transporte/metabolismo , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidases/metabolismo , Nanotubos de Carbono/química , Fibrose Pulmonar/patologia , Animais , Catepsina B/metabolismo , Linhagem Celular , Grupo dos Citocromos b/metabolismo , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Pulmão/patologia , Lisossomos/metabolismo , Macrófagos/metabolismo , Masculino , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 1 , Proteína 3 que Contém Domínio de Pirina da Família NLR , Estresse Oxidativo , Oxigênio/química , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória , Prata/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA