Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genes Dis ; 10(6): 2479-2490, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37554202

RESUMO

Post-translational modifications (PTM) are covalent modifications of proteins or peptides caused by proteolytic cleavage or the attachment of moieties to one or more amino acids. PTMs play essential roles in biological function and regulation and have been linked with several diseases. Modifications of protein acylation (Kac), a type of PTM, are known to induce epigenetic regulatory processes that promote various diseases. Thus, an increasing number of studies focusing on acylation modifications are being undertaken. Butyrylation (Kbu) is a new acylation process found in animals and plants. Kbu has been recently linked to the onset and progression of several diseases, such as cancer, cardiovascular diseases, diabetes, and vascular dementia. Moreover, the mode of action of certain drugs used in the treatment of lymphoma and colon cancer is based on the regulation of butyrylation levels, suggesting that butyrylation may play a therapeutic role in these diseases. In addition, butyrylation is also commonly involved in rice gene expression and thus plays an important role in the growth, development, and metabolism of rice. The tools and analytical methods that could be utilized for the prediction and detection of lysine butyrylation have also been investigated. This study reviews the potential role of histone Kbu, as well as the mechanisms underlying this process. It also summarizes various enzymes and analytical methods associated with Kbu, with the goal of providing new insights into the role of Kbu in gene regulation and diseases.

2.
FASEB J ; 36(10): e22553, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36111980

RESUMO

Mesenchymal stromal cells (MSCs) are attractive candidates for treating hepatic disorders given their potential to enhance liver regeneration and function. The paracrine paradigm may be involved in the mechanism of MSC-based therapy, and exosomes (Exo) play an important role in this paracrine activity. Hypoxia significantly improves the effectiveness of MSC transplantation. However, whether hypoxia preconditioned MSCs (Hp-MSCs) can enhance liver regeneration, and whether this enhancement is mediated by Exo, are unknown. In this study, mouse bone marrow-derived MSCs (BM-MSCs) and secreted Exo were injected through the tail vein. We report that Hp-MSCs promote liver regeneration after partial hepatectomy in mice through their secreted exosomes. Interestingly, MSC-Exo were concentrated in liver 6 h after administration and mainly taken up by macrophages, but not hepatocytes. Compared with normoxic MSC-Exo (N-Exo), hypoxic MSC-Exo (Hp-Exo) enhanced M2 macrophage polarization both in vivo and in vitro. Microarray analysis revealed significant enrichment of microRNA (miR)-182-5p in Hp-Exo compared with that in N-Exo. In addition, miR-182-5p knockdown partially abolished the beneficial effect of Hp-Exo. Finally, Hp-MSC-derived exosomal miR-182-5p inhibited theprotein expression of forkhead box transcription factor 1 (FOXO1) in macrophages, which inhibited toll-like receptor 4 (TLR4) expression and subsequently induced an anti-inflammatory response. These results highlight the therapeutic potential of Hp-Exo in liver regeneration and suggest that miR-182-5p from Hp-Exo facilitates macrophage polarization during liver regeneration by modulating the FOXO1/TLR4 signaling pathway.


Assuntos
Regeneração Hepática , Macrófagos , Células-Tronco Mesenquimais , MicroRNAs , Animais , Medula Óssea/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Hipóxia/metabolismo , Regeneração Hepática/genética , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , MicroRNAs/metabolismo , Receptor 4 Toll-Like/metabolismo
3.
J Nanobiotechnology ; 20(1): 289, 2022 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-35717234

RESUMO

Inorganic nanoparticles (INPs) have been paid great attention in the field of oncology in recent past years since they have enormous potential in drug delivery, gene delivery, photodynamic therapy (PDT), photothermal therapy (PTT), bio-imaging, driven motion, etc. To overcome the innate limitations of the conventional INPs, such as fast elimination by the immune system, low accumulation in tumor sites, and severe toxicity to the organism, great efforts have recently been made to modify naked INPs, facilitating their clinical application. Taking inspiration from nature, considerable researchers have exploited cell membrane-camouflaged INPs (CMCINPs) by coating various cell membranes onto INPs. CMCINPs naturally inherit the surface adhesive molecules, receptors, and functional proteins from the original cell membrane, making them versatile as the natural cells. In order to give a timely and representative review on this rapidly developing research subject, we highlighted recent advances in CMCINPs with superior unique merits of various INPs and natural cell membranes for cancer therapy applications. The opportunity and obstacles of CMCINPs for clinical translation were also discussed. The review is expected to assist researchers in better eliciting the effect of CMCINPs for the management of tumors and may catalyze breakthroughs in this area.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Fotoquimioterapia , Membrana Celular , Humanos , Hipertermia Induzida/métodos , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fototerapia/métodos
4.
Cancer Res ; 81(15): 4027-4040, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33985974

RESUMO

Triple-negative breast cancer (TNBC) exhibits a high mortality rate and is the most aggressive subtype of breast cancer. As previous studies have shown that histone deacetylases (HDAC) may represent molecular targets for TNBC treatment, we screened a small library of synthetic molecules and identified a potent HDAC inhibitor (HDACi), YF438, which exerts effective anti-TNBC activity both in vitro and in vivo. Proteomic and biochemical studies revealed that YF438 significantly downregulated mouse double minute 2 homolog (MDM2) expression. In parallel, loss of MDM2 expression or blocking MDM2 E3 ligase activity rendered TNBC cells less sensitive to YF438 treatment, revealing an essential role of MDM2 E3 ligase activity in YF438-induced inhibition of TNBC. Mechanistically, YF438 disturbed the interaction between HDAC1 and MDM2, induced the dissociation of MDM2-MDMX, and subsequently increased MDM2 self-ubiquitination to accelerate its degradation, which ultimately inhibited growth and metastasis of TNBC cells. In addition, analysis of clinical tissue samples demonstrated high expression levels of MDM2 in TNBC, and MDM2 protein levels closely correlated with TNBC progression and metastasis. Collectively, these findings show that MDM2 plays an essential role in TNBC progression and targeting the HDAC1-MDM2-MDMX signaling axis with YF438 may provide a promising therapeutic option for TNBC. Furthermore, this novel underlying mechanism of a hydroxamate-based HDACi in altering MDM2 highlights the need for further development of HDACi for TNBC treatment. SIGNIFICANCE: This study uncovers the essential role of MDM2 in TNBC progression and suggests that targeting the HDAC1-MDM2-MDMX axis with a hydroxamate-based HDACi could be a promising therapeutic strategy for TNBC.


Assuntos
Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Animais , Feminino , Humanos , Camundongos , Transfecção
6.
Nanoscale ; 12(47): 24030-24043, 2020 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-33291128

RESUMO

The metabolic enzyme-based arginine deprivation represents a tremendous opportunity to treat argininosuccinate synthetase (ASS1)-deficient tumors. Arginine deiminase (ADI), a typical representative, has aroused great interest. To date, the functional modification of ADI, such as PEGylation, has been applied to improve its weakness significantly, reducing its immunogenicity and extending its blood circulation time. However, the advantages of ADI, such as the cellular non-uptake property, are often deprived by current modification methods. The cellular non-uptake property of ADI only renders extracellular arginine degradation that negligibly influences normal cells. However, current-functionalized ADIs can be readily phagocytized by cells, causing the imbalance of intracellular amino acids and the consequent damage to normal cells. Therefore, it is necessary to exploit a new method that can simultaneously improve the weakness of ADI and maintain its advantage of cellular non-uptake. Here, we utilized a kind of phosphorylcholine (PC)-rich nanocapsule to load ADI. These nanocapsules possessed extremely weak cellular interaction and could avoid uptake by endothelial cells (HUVEC), immune cells (RAW 264.7), and tumor cells (H22), selectively depriving extracellular arginine. Besides, these nanocapsules increased the blood half-life time of ADI from the initial 2 h to 90 h and efficiently avoided its immune or inflammatory responses. After a single injection of ADI nanocapsules into H22 tumor-bearing mice, tumors were stably suppressed for 25 d without any detectable side effects. This new strategy first realizes the selective extracellular arginine deprivation for the treatment of ASS1-deficient tumors, potentially promoting the clinical translation of metabolic enzyme-based amino acid deprivation therapy. Furthermore, the research reminds us that the functionalization of drugs can not only improve its weakness but also maintain its advantages.


Assuntos
Arginina , Nanocápsulas , Animais , Linhagem Celular Tumoral , Células Endoteliais , Hidrolases , Camundongos , Polietilenoglicóis
7.
Theranostics ; 10(17): 7889-7905, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32685027

RESUMO

Rationale: Developing an effective nanoplatform to realize 'multi-in-one' is essential to broaden the therapeutic potential of combination therapy. Exosomes are ideal candidates since their intrinsic abilities of integrating multiple contents and functions. However, only limited efforts have been devoted to engineering exosomes to integrate the needed properties, also considering the safety and yield, for tumor-targeted and efficient gene/chemo combination therapy. Methods: Herein, by manipulating the exosome membrane, blood exosomes with high abundance and safety are engineered as a versatile combinatorial delivery system, where the doxorubicin (Dox) and cholesterol-modified miRNA21 inhibitor (miR-21i) are co-embedded into the lipid bilayer of exosomes, and the magnetic molecules and endosomolytic peptides L17E are bind to the exosome membrane through ligand-receptor coupling and electrostatic interactions, respectively. Results: It is proved that such engineering strategy not only preserves their intrinsic features, but also readily integrates multiple properties of tumor targeting, efficient transfection and gene/chemo combination therapy into blood exosomes. The lipid bilayer structure of exosomes allows them to co-load Dox and miR-21i with high-payloads. Moreover, profiting from the integration of magnetic molecules and L17E peptides, the engineered exosomes exhibit an enhanced tumor accumulation and an improved endosome escape ability, thereby specifically and efficiently delivering encapsulated cargos to tumor cells. As a result, a remarkable inhibition of tumor growth is observed in the tumor-bearing mice, and without noticeable side effects. Conclusions: This study demonstrates the potential of engineered blood exosomes as feasible co-delivery nanosystem for tumor-targeted and efficient combination therapy. Further development by replacing the drugs combined regimens can potentially make this engineered exosome become a general platform for the design of safe and effective combination therapy modality.


Assuntos
Engenharia Química/métodos , Sistemas de Liberação de Medicamentos/métodos , Exossomos/química , Terapia Genética/métodos , Neoplasias/terapia , Animais , Linhagem Celular Tumoral , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética/efeitos adversos , Humanos , Nanopartículas Magnéticas de Óxido de Ferro , Camundongos , MicroRNAs/antagonistas & inibidores , Neoplasias/genética , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Orthop Translat ; 24: 76-87, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32695607

RESUMO

OBJECTIVE: The healing of osteoporotic fractures in the elderly patients is a difficult clinical problem. Currently, based on the internal fixation of fractures, the available drug treatments mainly focus on either inhibiting osteoclast function, such as bisphosphonate, calcitonin, oestrogen or promoting osteogenesis, such as parathyroid hormones. However, the availability of current antiosteoporotic drugs in promoting osteoporotic fracture healing is limited. The objective of the present study was to investigate the ability of the MiR-21/nanocapsule to enhance the early bone repair of osteoporotic fractures. METHODS: Based on the presence of matrix metalloproteinases that are overexpressed at the fracture site, we designed the matrix metalloproteinase-sensitive nanocapsules which were formed by in situ free radical polymerisation on the surface of MiR-21 with 2-(methacryloyloxy) ethyl phosphorylcholine and the bisacryloylated VPLGVRTK peptide. The MiR-21/nanocapsule [n (miR-21)] and O-carboxymethyl chitosan (CMCS) were mixed until they formed a gel-like material [CMCS/n (miR-21)] with good fluidity and injectability. Thirty elderly Sprague Dawley (SD) rats (female, 14-month-old, 380 ± 10 g) were subjected to bilateral removal of the ovaries (ovariectomised). All rats were subjected to bilateral bone defects (2 mm diameter) of the proximal tibia and randomly divided into three groups (groups A, B, and C): separately injected with CMCS/n (miR-21), CMCS/n (NC-miR), and saline. Micro-computed tomography (CT) imaging was performed to evaluate newly formed bone volume and connectivity. Nondecalcified histology and toluidine blue staining were performed to measure the effects of CMCS/n (miR-21) on bone repair. In vitro, the effect of n (miR-21) on osteogenic differentiation to bone marrow mesenchymal stem cells (BMSCs) which derived from the ovariectomised rat model was observed. RESULTS: The morphology of n (miR-21) was a regular spherical nanocapsule with a uniform small size (25-35 nm). The results confirmed that n (miR-21) could be efficiently phagocytosed by BMSCs and released in the cytoplasm to promote osteogenesis. The expression level of alkaline phosphatase and Runt-related transcription factor 2 mRNA in the n (miR-21) group was higher than that in the n (NC-miR) group. Animal experiments proved that CMCS/n (miR-21) produced better bone repair compared with the CMCS/n (NC-miR) group in the early stages of fracture healing at 4 weeks. In the late stage of fracture healing (8 weeks), micro-CT quantitative analysis showed that the new bone trabeculae in the CMCS/n (miR-21) group has decreased compared with the CMCS/n (NC-miR) group. In the CMCS/n (miR-21) group, the new cancellous bone had been absorbed, and the process of bone healing was almost completed. In contrast, the new bone in the CMCS/n (NC-miR) and the control groups was still in the healing process. CONCLUSION: The cytological tests confirmed that n (miR-21) can promote osteogenic differentiation of BMSCs derived from the osteoporosis rat model. Furthermore, the results of animal tests demonstrated that local injection of CMCS/n (miR-21) promoted the early healing of osteoporotic bone defects. Consequently CMCS/n (miR-21) promoted the bone repair process to enter the moulding phase earlier. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: CMCS/n (miR-21) can be widely applied to elderly patients with osteoporotic fractures. This method can help patients with osteoporotic fractures recover earlier and avoid serious complications. It provides a potential approach for the clinical treatment of osteoporotic fractures in the elderly.

9.
Cancer Biol Med ; 17(2): 433-443, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32587779

RESUMO

Objective: The introduction of therapeutic antibodies (tAbs) into clinical practice has revolutionized tumor treatment strategies, but their tumor therapy efficiency is still far below expectations because of the rapid degradation and limited tumor accumulation of tAbs. Methods: We developed a nanocapsule-based delivery system to induce the self-augmentation of the enhanced permeability and retention (EPR) effect. This system constantly penetrated across the blood-tumor barrier into the tumor while avoiding the attack of tAbs by the immune system. The biodistribution and therapeutic effect were tested with single dose administration of nanocapsule-tAbs in vivo. Results: The accumulation of Nano(cetuximab) within subcutaneous PC9 tumors was gradually enhanced over 6 days after single dose administration, which was contrary to the biodistribution of native cetuximab. Nano(cetuximab) accumulated in tumor tissues via the EPR effect and released cetuximab. The released cetuximab acted on vascular endothelial cells to destroy the blood-tumor barrier and induce self-augmentation of the EPR effect, which in turn contributed to further tumor accumulation of long-circulating Nano(cetuximab). Compared with single dose administration of native cetuximab, Nano(cetuximab) showed an effective tumor suppressive effect for 3 weeks. Conclusions: The nanocapsule-based delivery system efficiently delivered tAbs to tumor tissues and released them to boost the EPR effect, which facilitated further tumor accumulation of the tAbs. This novel self-augmentation of the EPR effect facilitated by the biological characteristics of tAbs and nanotechnology contributed to the improvement of the therapeutic effect of tAbs, and stimulated new ideas for antibody-based tumor therapy.


Assuntos
Cetuximab/farmacologia , Células Endoteliais/efeitos dos fármacos , Nanocápsulas , Animais , Sistemas de Liberação de Medicamentos , Feminino , Camundongos , Camundongos Nus , Permeabilidade , Células RAW 264.7 , Distribuição Tecidual
10.
Biosci Rep ; 40(3)2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32124924

RESUMO

The use of circulating microRNAs as biomarkers opens up new opportunities for the diagnosis of cardiovascular diseases because of their specific expression profiles. The aim of the present study was to identify circulating microRNAs in human plasma as potential biomarkers of heart failure and related diseases. We used real-time quantitative PCR to screen microRNA in plasma samples from 62 normal controls and 62 heart failure samples. We found that circulating miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 expressed differently between healthy controls and heart failure patients. Plasma levels of miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 were unaffected by hemolysis. Correlation analysis showed any two of these miRNAs possess a strong correlation, indicating a possibility of combined analysis. MiR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 could be combined in two or three or more combinations. The results suggest that miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 may be a new diagnostic biomarker for heart failure and related diseases.


Assuntos
Perfilação da Expressão Gênica/métodos , Insuficiência Cardíaca/genética , MicroRNAs/genética , Idoso , Biomarcadores Tumorais/genética , China , Feminino , Insuficiência Cardíaca/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real/métodos
11.
J Cell Mol Med ; 24(1): 1146-1150, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31709737

RESUMO

Coronary artery disease (CAD) is one of the biggest threats to human life. Circulating microRNAs (miRNAs) have been reported to be linked to the pathogenesis of CAD, indicating the possible role in CAD diagnosis. The present study aimed to explore the expression profile of plasma miRNAs and estimate their value in diagnosis for CAD. 67 Non-CAD control subjects and 88 CAD patients were enrolled. We conducted careful evaluation by RT-PCR analysis, Spearman rank correlation coefficients analysis, Receiver Operating Characteristic (ROC) curves analysis and so on. The plasma levels of six miRNAs known to be related to CAD were measured and three of them showed obvious expression change. Circulating miR-29a-3p, miR-574-3p and miR-574-5p were all significantly increased. ROC analysis revealed the probability of the three miRNAs as biomarkers with AUCs (areas under the ROC curve) of 0.830, 0.792 and 0.789, respectively. They were significantly correlated with each other in CAD patients, suggesting the possibility of joint diagnosis. The combined AUC was 0.915, much higher than each single miRNA. Therefore, our study revealed three promising biomarkers for early diagnosis of CAD. The combination of these miRNAs may act more effectively than individual ones for CAD diagnosis.


Assuntos
Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , MicroRNA Circulante/sangue , MicroRNA Circulante/genética , Doença da Artéria Coronariana/diagnóstico , Regulação Neoplásica da Expressão Gênica , Estudos de Casos e Controles , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/genética , Feminino , Seguimentos , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Curva ROC
12.
Theranostics ; 9(25): 7616-7627, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695790

RESUMO

Cell-based immunotherapy for the treatment of hematologic malignancies, such as leukemia and lymphoma, has seen much success and played an increasingly important role in clinical studies. Nevertheless, the efficacy of immunotherapy in solid tumors still needs improvements due to the immunosuppressive properties of tumor cells and the microenvironment. To overcome these limitations, we prepared a novel tumor-targeting delivery system based on the underlying mechanism of immune-targeted cell death that encapsulated granzyme B protein within a porous polymeric nanocapsule. Methods: A cell-penetrating peptide TAT was attached onto granzyme B (GrB) to enhance its transmembrane transport efficiency and potency to induce cell apoptosis. The endocytosis and internalization pathways of GrB-TAT (GrB-T) were analyzed in comparison with perforin by confocal microscopy and flow cytometry. Furthermore, the positively charged GrB-T was wrapped into nanoparticles by p-2-methacryloyloxy ethyl phosphorylcholine (PMPC)-modified HA (hyaluronic acid). The nanoparticles (called TCiGNPs) were characterized in terms of zeta potential and by transmission electron microscopy (TEM). The in vitro anti-tumor effects of GrB-T were examined by cell apoptosis assay and Western blotting analysis. The in vivo anti-tumor therapeutic efficacy of TCiGNPs was evaluated in a mouse tumor model. Results: The TAT peptide could play a role similar to perforin to mediate direct transmembrane transfer of GrB and improve GrB-induced cell apoptosis. The TCiGNPs were successfully synthesized and accumulated in the solid tumor through enhanced permeability and retention (EPR) effect. In the tumor microenvironment, TCiGNPs could be degraded by hyaluronidase and triggered the release of GrB-T. The TAT peptide enabled the translocation of GrB across the plasma membrane to induce tumor cell apoptosis in vivo.Conclusion: We successfully developed a granzyme B delivery system with a GrB-T core and a PMPC/HA shell that simulated CTL/NK cell-mediated cancer immunotherapy mechanism. The GrB delivery system holds great promise for cancer treatment analogous to the CTL/NK cell-induced immunotherapy.


Assuntos
Granzimas/administração & dosagem , Sistema Imunitário/efeitos dos fármacos , Nanopartículas/administração & dosagem , Neoplasias/imunologia , Neoplasias/terapia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Peptídeos Penetradores de Células/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Humanos , Imunoterapia/métodos , Células Matadoras Naturais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Perforina/administração & dosagem , Linfócitos T Citotóxicos/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
13.
Theranostics ; 9(25): 7680-7696, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695794

RESUMO

Blood transferrin receptor-positive (TfR+) exosomes are a kind of optimized drug delivery vector compared with other kinds of exosomes due to their easy access and high bio-safety. Their application facilitates the translation from bench to bedside of exosome-based delivery vehicles. Methods: In this study, a pH-responsive superparamagnetic nanoparticles cluster (denoted as SMNC)-based method was developed for the precise and mild separation of blood TfR+ exosomes. Briefly, multiple superparamagnetic nanoparticles (SPMNs) labeled with transferrins (Tfs) could precisely bind to blood TfR+ exosomes to form an exosome-based cluster due to the specific recognition of TfR by Tf. They could realize the precise magnetic separation of blood TfR+ exosomes. More importantly, the pH-responsive dissociation characteristic of Tf and TfR led to the mild collapse of clusters to obtain pure blood TfR+ exosomes. Results: Blood TfR+ exosomes with high purity and in their original state were successfully obtained through the pH-responsive SMNC-based method. These can load Doxorubicin (DOX) with a loading capacity of ~10% and dramatically increase the tumor accumulation of DOX in tumor-bearing mice because of their innate passive-targeting ability. In addition, blood TfR+ exosomes changed the biodistribution of DOX leading to the reduction of side effects. Compared with free DOX, DOX-loaded blood TfR+ exosomes showed much better tumor inhibition effects on tumor-bearing mice. Conclusion: Taking advantage of the pH-responsive binding and disaggregation characteristics of Tf and TfR, the SMNC-based method can precisely separate blood TfR+ exosomes with high purity and in their original state. The resulting blood TfR+ exosomes showed excellent bio-safety and enable the efficient delivery of chemotherapeutics to tumors, facilitating the clinical translation of exosome-based drug delivery systems.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/química , Exossomos/química , Neoplasias/tratamento farmacológico , Receptores da Transferrina/sangue , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Sistemas de Liberação de Medicamentos/métodos , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Distribuição Tecidual
14.
Adv Mater ; 31(19): e1805697, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30773720

RESUMO

As an essential component of immunotherapy, monoclonal antibodies (mAbs) have emerged as a class of powerful therapeutics for treatment of a broad range of diseases. For central nervous system (CNS) diseases, however, the efficacy remains limited due to their inability to enter the CNS. A platform technology is reported here that enables effective delivery of mAbs to the CNS for brain tumor therapy. This is achieved by encapsulating the mAbs within nanocapsules that contain choline and acetylcholine analogues; such analogues facilitate the penetration of the nanocapsules through the brain-blood barrier and the delivery of mAbs to tumor sites. This platform technology uncages the therapeutic power of mAbs for various CNS diseases that remain poorly treated.


Assuntos
Anticorpos Monoclonais/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/tratamento farmacológico , Sistema Nervoso Central/efeitos dos fármacos , Nanocápsulas/química , Acetilcolina/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo , Colina/metabolismo , Reagentes de Ligações Cruzadas/química , Liberação Controlada de Fármacos , Humanos , Imunoterapia/métodos , Masculino , Camundongos Endogâmicos BALB C , Tamanho da Partícula , Permeabilidade , Fosforilcolina/química
15.
Theranostics ; 8(6): 1540-1557, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29556340

RESUMO

Exosomes play critical roles in intercellular communication in both nearby and distant cells in individuals and organs. Polymerase I and transcript release factor (PTRF), also known as Cavin1, has previously been described as a critical factor in caveola formation, and aberrant PTRF expression has been reported in various malignancies. However, the function of PTRF in tumor progression remains controversial, and its role in glioma is poorly understood. In this study, we report that PTRF is associated with malignancy grade and poor prognosis in glioma patients. Our previous study using two proteomics methods, tandem mass tag (TMT) and data-independent acquisition (DIA), showed that EGFRvIII overexpression increased PTRF expression at the protein level. In contrast, blocking PI3K and AKT using LY294002 and MK-2206, respectively, decreased PTRF expression, showing that PTRF is regulated in the EGFR/PI3K/AKT pathway. ChIP-PCR analysis showed that PTRF is transcriptionally regulated by the H3K4me3 and H3K27me3 modifications. Furthermore, PTRF overexpression increased exosome secretion and induced cell growth in vitro. More importantly, overexpressing PTRF induced the malignancy of nearby cells in vivo, suggesting that PTRF alters the microenvironment through intercellular communication via exosomes. Furthermore, analysis of clinical samples showed a positive correlation between tumor grade and PTRF expression in both tumor tissues and exosomes isolated from blood harvested from glioma patients, and PTRF expression in exosomes isolated from the sera of GBM patients was decreased after surgery. In conclusion, PTRF serves as a promising biomarker in both tumor samples and serum exosomes, thus facilitating the detection of glioma and potentially serving as a therapeutic target for glioblastoma multiforme.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Exossomos/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteínas de Ligação a RNA/genética , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Exossomos/patologia , Feminino , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Histonas/genética , Histonas/metabolismo , Humanos , Camundongos Nus , Gradação de Tumores , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Prognóstico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Mater Chem B ; 6(18): 2758-2768, 2018 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32254228

RESUMO

Exosomes, a class of natural nanoparticles, have recently been used for drug delivery. To move from bench to bedside, exosomes should meet the "STOP" criteria: (1) high Safety; (2) good Targeting ability; (3) rapid Obtainment; and (4) high Purity. To the best of our knowledge, however, no studies on exosomes that can satisfy all the abovementioned criteria have been reported to date. In this study, an optimized superparamagnetic nanoparticle cluster (SMNC)-based method was used to easily achieve the abovementioned objective. Multiple superparamagnetic nanoparticles (SPMNs) labeled by holo-transferrin with PEG spacers anchor onto an exosome to form an upgraded exosome-based superparamagnetic nanoparticle cluster (denoted SMNC-EXO-PLUS). SMNC-EXO-PLUSs separated exosomes from healthy animal blood to ensure high safety, and their formation based on the specific binding of SPMNs to exosomes endowed exosomes with high purity. In addition, SMNC-EXO-PLUSs possessed strong magnetic response, realizing their rapid obtainment and good targeting ability. The results demonstrated that the cell viability was maintained as high as 88% in a 1000 µg mL-1 SMNC-EXO-PLUS solution, and the time interval from the extraction of blood to the acquisition of purified exosomes could be reduced to 3.5 h. Furthermore, the effects of in vivo tumor inhibition of drug-loaded SMNC-EXO-PLUSs (denoted as D-SMNC-EXO-PLUSs) were pronounced. Based on the "STOP" criteria, we exploited a new exosome-related drug delivery vehicle, dramatically promoting the clinical translation of exosomes.

17.
J Mater Chem B ; 6(20): 3331-3339, 2018 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32254390

RESUMO

The limited penetration of drug delivery vehicles in tumors reduces their therapeutic efficacy, which is a major obstacle. Thus, great efforts have already been paid to regulate both tumor tissue and the physicochemical properties of drug delivery vehicles to achieve deeper penetration. However, the principles for regulating these properties remain unclear. Herein, we investigated the performance of nanocapsules in different tumor stages, and particularly focused on the difference in the performance of nanocapsules with different surface charges. Our in vivo and in vitro results showed that when the tumor was small in the early stage, the electropositive nanocapsules could penetrate much deeper than the electronegative nanocapsules; however, when the tumor was quite large in the late stage, the electronegative nanocapsules would penetrate further into the tissue. This study proposes a potential strategy for tumor therapy by selecting drug delivery vehicles with an appropriate surface charge based on the tumor development stages.

18.
ACS Nano ; 10(3): 3323-33, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26938862

RESUMO

Exosomes are a class of naturally occurring nanoparticles that are secreted endogenously by mammalian cells. Clinical applications for exosomes remain a challenge because of their unsuitable donors, low scalability, and insufficient targeting ability. In this study, we developed a dual-functional exosome-based superparamagnetic nanoparticle cluster as a targeted drug delivery vehicle for cancer therapy. The resulting exosome-based drug delivery vehicle exhibits superparamagnetic behavior at room temperature, with a stronger response to an external magnetic field than individual superparamagnetic nanoparticles. These properties enable exosomes to be separated from the blood and to target diseased cells. In vivo studies using murine hepatoma 22 subcutaneous cancer cells showed that drug-loaded exosome-based vehicle delivery enhanced cancer targeting under an external magnetic field and suppressed tumor growth. Our developments overcome major barriers to the utility of exosomes for cancer application.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Carcinoma Hepatocelular/tratamento farmacológico , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Exossomos/química , Neoplasias Hepáticas/tratamento farmacológico , Imãs/química , Nanopartículas/química , Animais , Antibióticos Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/patologia , Campos Magnéticos , Camundongos , Nanopartículas/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA