Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Small ; 19(24): e2207315, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36929209

RESUMO

Polyoxometalates (POMs) are widely used in catalysis, energy storage, biomedicine, and other research fields due to their unique acidity, photothermal, and redox features. However, the leaching and agglomeration problems of POMs greatly limit their practical applications. Confining POMs in a host material is an efficient tool to address the above-mentioned issues. POM@host materials have received extensive attention in recent years. They not only inherent characteristics of POMs and host, but also play a significant synergistic effect from each component. This review focuses on the recent advances in the development and applications of POM@host materials. Different types of host materials are elaborated in detail, including tubular, layered, and porous materials. Variations in the structures and properties of POMs and hosts before and after confinement are highlighted as well. In addition, an overview of applications for the representative POM@host materials in electrochemical, catalytic, and biological fields is provided. Finally, the challenges and future perspectives of POM@host composites are discussed.

2.
Acc Chem Res ; 52(7): 1750-1760, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31243966

RESUMO

Over the past decades, major efforts were undertaken to develop devices on a nanoscale level for the efficient and nontoxic delivery of molecules to tissues and cells, for the purpose of either diagnosis or treatment of disease. The application of such devices in drug delivery has proven to be beneficial for matters as diverse as drug solubility, drug targeting, controlled drug release, and transport of drugs across cellular barriers. Multiple nanotherapeutics have been approved for clinical treatment, and more products are being evaluated in preclinical and clinical trials. However, many biological barriers hinder the medical application of nanocarriers. There are two main classes of barriers that need to be overcome by drug nanocarriers: extracellular and intracellular barriers, both of which may capture and/or destroy therapeutics before they reach their target site. This Account discusses major biological barriers that are confronted by nanotherapeutics, following their systemic administration, focusing on cellular entry and endosomal escape of gene delivery vectors. The use of pH-responsive materials to overcome the endosomal barrier is addressed. Historically, cell biologists have studied the interaction between cells and pathogens in order to unveil the mechanisms of endocytosis and cell signaling. Meanwhile, it is becoming clear that cells may respond in similar ways to artificial drug delivery systems and, consequently, that knowledge on the cellular response against both pathogens and nanoparticulate systems will aid in the design of improved nanomedicine. A close collaboration between bioengineers and cell biologists will promote this development. At the same time, we have come to realize that tools that we use to study fundamental cellular processes, including metabolic inhibitors of endocytosis and overexpression/downregulation of proteins, may cause changes in cellular physiology. This calls for the implementation of refined methods to study nanocarrier-cell interactions, as is discussed in this Account. Finally, recent papers on the dynamics of cargo release from endosomes by means of live cell imaging have significantly advanced our understanding of the transfection process. They have initiated discussion (among others) on the limited number of endosomal escape events in transfection, and on the endosomal stage at which genetic cargo is most efficiently released. Advancements in imaging techniques, including super-resolution microscopy, in concert with techniques to label endogenous proteins and/or label proteins with synthetic fluorophores, will contribute to a more detailed understanding of nanocarrier-cell dynamics, which is imperative for the development of safe and efficient nanomedicine.


Assuntos
Portadores de Fármacos/metabolismo , Endocitose/fisiologia , Endossomos/metabolismo , Lipossomos/metabolismo , Polímeros/metabolismo , DNA/metabolismo , Portadores de Fármacos/química , Técnicas de Transferência de Genes , Células HeLa , Humanos , Lipossomos/química , Nanopartículas/química , Nanopartículas/metabolismo , Polímeros/química , RNA/metabolismo
3.
Mol Pharm ; 15(8): 3032-3045, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29939755

RESUMO

It is not efficient enough using the current approaches for tumor-selective drug delivery based on the EPR effect and ligand-receptor interactions, and they have largely failed to translate into the clinic. Therefore, it is urgent to explore an enhanced strategy for effective delivery of anticancer agents. Clinically, many cancers require large amounts of glutamine for their continued growth and survival, resulting in circulating glutamine extraction by the tumor being much greater than that for any organs, behaving as a "glutamine trap". In the present study, we sought to elucidate whether the glutamine-trap effect could be exploited to deliver therapeutic agents to selectively kill cancer cells. Here, a macromolecular glutamine analogue, glutamine-functionalized branched polyethylenimine (GPI), was constructed as the carrier to deliver anti-CD47 siRNA for the blockage of CD47 "don't eat me" signals on cancer cells. The GPI/siRNA glutamine-rich polyplexes exhibited remarkably high levels of cellular uptake by glutamine-dependent lung cancer cells, wild-type A549 cells (A549WT), and its cisplatin-resistant cells (A549DDP), specifically under glutamine-depleted conditions. It was noted that the glutamine transporter ASCT2 was highly expressed both on A549WT and A549DDP but with almost no expression in normal human lung fibroblasts cells. Inhibition of ASCT2 significantly prevented the internalization of GPI polyplexes. These findings raised the intriguing possibility that the glutamine-rich GPI polyplexes utilize the ASCT2 pathway to selectively facilitate their cellular uptake by cancer cells. GPI further delivered anti-CD47 siRNA efficiently both in vitro and in vivo to downregulate the intratumoral mRNA and protein expression levels of CD47. CD47 functions as a "don't eat me" signal and binds to the immunoreceptor SIRPα inducing evasion of phagocytic clearance. GPI/anti-CD47 siRNA polyplexes achieved significant antitumor activities both on A549WT and A549DDP tumor-bearing nude mice. Notably, it had no adverse effect on CD47-expressing red blood cells and platelets, likely because of selective delivery. Therefore, the glutamine-rich carrier GPI driven by the glutamine-trap effect provides a promising new strategy for designing anticancer drug delivery systems.


Assuntos
Antígeno CD47/antagonistas & inibidores , Portadores de Fármacos/química , Neoplasias Pulmonares/tratamento farmacológico , RNA Interferente Pequeno/administração & dosagem , Células A549 , Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Sistema ASC de Transporte de Aminoácidos/metabolismo , Animais , Antígeno CD47/genética , Dipeptídeos/farmacologia , Fibroblastos , Glutamina/análogos & derivados , Glutamina/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Antígenos de Histocompatibilidade Menor/metabolismo , Polietilenoimina/química , RNA Interferente Pequeno/genética , Cloridrato de Raloxifeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Zhongguo Yi Liao Qi Xie Za Zhi ; 42(1): 74-76, 2018 Jan 30.
Artigo em Zh | MEDLINE | ID: mdl-29862755

RESUMO

Hyaluronic acid is a linear, high molecular weight glycosaminoglycan. It has the characteristics of good biocompatibility, hydrophilicity and antigenicity. In this paper, the application of hyaluronic acid in the fields of ophthalmology, surgery, arthritis, cosmetic surgery and trauma repair is reviewed as well as the potential application in the field of tissue engineering.


Assuntos
Ácido Hialurônico/uso terapêutico , Engenharia Tecidual , Cicatrização , Humanos
5.
AAPS PharmSciTech ; 18(6): 2329-2335, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28116599

RESUMO

Most therapeutic proteins are classified as class III drugs according to the Biopharmaceutical Classification System means that the low permeability across the intestinal epithelium is the rate-limited step for their oral absorption. Cationic chitosan nanoparticles have been found to open the tight junctions between epithelial cells. On the other hand, bioadhesive delivery devices could prolong the gastrointestinal residence time. In the present study, we developed a novel nano-reservoir bioadhesive tablets that combining the advantages of cationic nanoparticles and bioadhesive delivery devices anticipated achieving effective transport of sufficient protein drugs across the intestinal epithelium. The nano-reservoir in bioadhesive tablets was composed of chitosan nanoparticles (CS-NPs) loading a model protein drug bovine serum albumin (BSA). The formula of bioadhesive tablets was optimized by using rotatable central composite design and response surface methodology. The nano-reservoir, BSA-loaded CS-NPs, had an average particle diameter of 312.5 ± 12.89 nm and zeta-potential value of 26.76 ± 3.56 mV. Carboxymethyl chitosan added to the formula significantly ameliorated the tight junction damage of the Caco-2 cell monolayer induced by CS-NPs, meanwhile maintained the high transport efficiency of BSA. Permeability study exhibited that these nano-reservoir bioadhesive tablets combining the advantages of cationic nanoparticles and bioadhesive tablets significantly enhanced BSA transport through rabbit small intestine in comparison with either conventional bioadhesive tablets or CS-NPs. Therefore, these nano-reservoir bioadhesive tablets provided a great potential dosage form design for the oral delivery of protein drugs.


Assuntos
Intestino Delgado/metabolismo , Soroalbumina Bovina/farmacocinética , Comprimidos/farmacocinética , Animais , Células CACO-2 , Bovinos , Quitosana/administração & dosagem , Quitosana/análogos & derivados , Humanos , Mucosa Intestinal/metabolismo , Nanopartículas/administração & dosagem , Permeabilidade , Coelhos
6.
Mater Today Bio ; 26: 101087, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38784443

RESUMO

Given multifunction of copper (Cu) contributing to all stages of the physiology of wound healing, Cu-based compounds have great therapeutic potentials to accelerate the wound healing, but they must be limited to a very low concentration range to avoid detrimental accumulation. Additionally, the cellular mechanism of Cu-based compounds participating the healing process remains elusive. In this study, copper oxide nanoparticles (CuONPs) were synthesized to mimic the multiple natural enzymes and trapped into PEG-b-PCL polymersomes (PS) to construct cupric-polymeric nanoreactors (CuO@PS) via a direct hydration method, thus allowing to compartmentalize Cu-based catalytic reactions in an isolated space to improve the efficiency, selectivity, recyclability as well as biocompatibility. While nanoreactors trafficked to lysosomes following endocytosis, the released Cu-based compounds in lysosomal lumen drove a cytosolic Cu+ influx to mobilize Cu metabolism mostly via Atox1-ATP7a/b-Lox axis, thereby activating the phosphorylation of mitogen-activated protein kinase 1 and 2 (MEK1/2) to initiate downstream signaling events associated with cell proliferation, migration and angiogenesis. Moreover, to facilitate to lay on wounds, cupric-polymeric nanoreactors were finely dispersed into a thermosensitive Pluronic F127 hydrogel to form a composite hydrogel sheet that promoted the healing of chronic wounds in diabetic rat models. Hence, cupric-polymeric nanoreactors represented an attractive translational strategy to harness cellular Cu metabolism for chronic wounds healing.

7.
J Thorac Dis ; 14(5): 1478-1487, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35693606

RESUMO

Background: This study aimed to summarize the available data on the association between the severity of (COVID-19) and routine blood indicators, inflammatory, biochemical parameters and coagulation parameter. Methods: A literature search was conducted of PubMed, EMBASE, and Web of Sciences, CNKI, WanFang database providing relevant data. Random-effects meta-analysis was used to pool effect sizes. Results: In patients with severe symptoms, interleukin-6, [IL-6; standardized mean difference (SMD) =1.15, 95% confidence interval (95% CI): 1.01, 1.29, P<0.001, n=1,121], interleukin-10 (IL-10; SMD =0.92, 95% CI: 0.75, 1.08, P<0.001, n=782), interleukin-4 (IL-4; SMD =0.2, 95% CI: 0.01, 0.39, P=0.04, n=500), procalcitonin (PCT; SMD =1.16, 95% CI: 0.99, 1.33, P<0.001, n=734), C-reactive protein (CRP; SMD =1.42, 95% CI: 1.27, 1.57, P<0.001, n=1,286), serum amyloid A (SAA; SMD =2.82, 95% CI: 2.53, 3.11, P<0.001, n=502) neutrophil count (SMD =0.63, 95% CI: 0.44, 0.82, P<0.001, n=558), alanine aminotransferase (ALT; SMD =2.72, 95% CI: 2.43, 3.02, P<0.001, n=538), aspartate aminotransferase (AST; SMD =2.75, 95% CI: 2.37, 3.12, P<0.001, n=313), lactate dehydrogenase (LDH; SMD =4.01, 95% CI: 3.79, 4.24, P<0.001, n=1,055), creatine kinase (CK; SMD =2.62, 95% CI: 2.2, 3.03, P<0.001, n=230;), CK-MB isoenzyme (CK-MB; SMD =3.07, 95% CI: 2.81, 3.34, P<0.001, n=600, activated partial thromboplastin time (APTT; SMD =0.63, 95% CI: 0.39, 0.87, P<0.001, n=351), and prothrombin time (P-T; SMD =1.83, 95% CI: 1.55, 2.11, P<0.001, n=351) were significantly higher than in patients with mild symptoms. On the contrary, lymphocyte count (SMD =-1.04, 95% CI: -1.21, -0.86, P<0.001, n=805) platelets (SMD =-1.47, 95% CI: -1.7, -1.24, P<0.001, n=653), monocyte count (SMD =-0.56, 95% CI: -0.8, -0.32, P<0.001, n=403), and albumin (SMD =-2.95, 95% CI: -3.21, -2.7, P<0.001, n=637) was significantly lower in patients with severe symptoms than in patients with mild symptoms. IL-6 (SMD =2.62, 95% CI: 2.15, 3.09, P<0.001, n=185), PCT (SMD =0.2, 95% CI: 0.16, 0.23, P<0.001, n=156), creatinine (SMD =2.29, 95% CI: 1.87, 2.7, P<0.001, n=213), and neutrophil counts (SMD =2.77, 95% CI: 2.38, 3.16, P<0.001, n=260) in patients with COVID-19 in the death group were significantly higher than that in patients in the survival group, while the lymphocyte count was significantly lower. Conclusions: In summary, current evidence show that those laboratory indicators are associated with the severity of COVID-19 and thus could be used as prognostic risk stratification of patients with COVID-19.

8.
Medicine (Baltimore) ; 101(51): e32340, 2022 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-36595789

RESUMO

BACKGROUND: The mammalian target of rapamycin (mTOR) has a crucial role in carcinogenesis, angiogenesis, cellular proliferation, and metastasis; however, its significance in non-small cell lung cancer (NSCLC) remains contentious. Consequently, this study aims to assess the clinicopathological and prognostic importance of mTOR/p-mTOR expression in NSCLC. METHODS: Literature retrieval was undertaken by searching English databases PubMed, EMBASE, Web of Science, and Cochrane Library as well as Chinese databases CNKI, Wan Fang, and VIP for full-text publications that satisfied our eligibility criteria up to November 2021. STATA 12.0 was used to conduct statistical analysis (STATA Corporation, College Station, TX). RESULTS: This meta-analysis includes a total of 4683 patients from 28 primary publications. mTOR/p-mTOR expression was associated with sex (OR = 0.608, 95% CI: 0.442-0.836), lymph node metastasis (OR = 2.084, 95% CI: 1.437-3.182), and CEA (OR = 1.584, 95% CI: 1.135-2.209), but not with age, histological type, depth of tumor invasion, distant metastasis, TNM stage, differentiation degree, tumor size, or smoking. In addition, the expression of mTOR/p-mTOR is related to shorter overall survival in NSCLC patients (HR = 1.415, 95% CI: 1.051-1.905). CONCLUSION: Positive mTOR/p-mTOR expression was substantially correlated with unfavorable conditions on the sex, lymph node metastases, and CEA levels. mTOR/p-mTOR may indicate a bad prognosis for NSCLC. The current findings must be confirmed and changed by other high-quality research employing a multivariate analysis on bigger sample size.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Prognóstico , Neoplasias Pulmonares/patologia , Metástase Linfática , Serina-Treonina Quinases TOR/metabolismo , Biomarcadores Tumorais/metabolismo
9.
Drug Deliv ; 29(1): 2414-2427, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35904177

RESUMO

Polymersomes possess the self-assembly vesicular structure similar to liposomes. Although a variety of comparisons between polymersomes and liposomes in the aspects of physical properties, preparation and applications have been elaborated in many studies, few focus on their differences in drug encapsulation, delivery and release in vitro and in vivo. In the present work, we have provided a modified direct hydration method to encapsulate anti-cancer drug paclitaxel (PTX) into PEG-b-PCL constituted polymersomes (PTX@PS). In addition to advantages including narrow particle size distribution, high colloid stability and moderate drug-loading efficiency, we find that the loaded drug aggregate in small clusters and reside through the polymersome membrane, representing a unique core-satellite structure which might facilitate the sustained drug release. Compared with commercial liposomal PTX formulation (Lipusu®), PTX@PS exhibited superb tumor cell killing ability underlain by multiple pro-apoptotic mechanisms. Moreover, endocytic process of PTX@PS significantly inhibits drug transporter P-gp expression which could be largely activated by free drug diffusion. In glioma mice models, it has also confirmed that PTX@PS remarkably eradicate tumors, which renders polymersomes as a promising alternative to liposomes as drug carriers in clinic.


Assuntos
Antineoplásicos , Lipossomos , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Camundongos , Paclitaxel/química , Polietilenoglicóis/química
10.
Pharm Res ; 28(4): 812-26, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21161337

RESUMO

PURPOSE: Inefficient endosomal escape and poor nuclear import are thought to contribute to low gene transfer efficiency of polycations. To overcome these drawbacks, we prepared multiple gene delivery formulations including low cytotoxic polycation, histone containing NLSs and chloroquine as the endosomolytic agent. METHODS: Comb-shaped poly (L-glutamic acid) grafted low-molecular-weight polyethylenimine (PLGE) copolymer was synthesized by aminolysis of poly-γ-benzyl-L-glutamate using low-molecular-weight polyethylenimine (800 Da). The formation of DNA/histone/PLGE terplex was observed by atomic force microscope and gel retardation assay. The particle size and zeta potential of DNA complexes with varying content of histone were also measured to confirm the terplex formation. Cytotoxicity of vectors was assayed by MTT. Multiple gene delivery formulations were optimized to their best transfection efficiency that was monitored by fluorescence microscope and flow cytometry. In vivo gene delivery of the optimal formulation was evaluated by the GFP-expression levels in drosophila melanogaster. RESULTS: The DNA/histone/PLGE terplex was successfully formed. The PLGE and histone together condensed DNA into small, discrete particles (less than 200 nm in diameter) in isotonic solution. Cytotoxicity of PLGE and histone were much lower than that of PEI 25 K. Either histone or chloroquine contributed to enhancing the levels of transfection activity of PLGE polymer. However, chloroquine and histone did not show a synergistic effect on the improvement of transfection efficiency. The optimal formulation was the DNA/histone/PLGE terplex at the N/P ratio of 15 and histone/ DNA weight ratio of 0.8. Compared with Lipofectamine 2000 and PEI 25 K, the optimal formulation showed significantly increased levels of GFP-expression both in vitro and in vivo. CONCLUSION: This formulation provided a versatile approach for preparing high efficiency of the polycation-based gene vectors. It also reinforced the finding of earlier studies that nuclear import and endosomal escape were rate-limiting steps for nonviral gene delivery.


Assuntos
DNA/administração & dosagem , Portadores de Fármacos/química , Técnicas de Transferência de Genes , Histonas/química , Polietilenoimina/química , Ácido Poliglutâmico/química , Animais , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cloroquina/química , DNA/genética , Drosophila melanogaster/genética , Portadores de Fármacos/efeitos adversos , Portadores de Fármacos/síntese química , Endossomos/metabolismo , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Estrutura Molecular , Peso Molecular , Sinais de Localização Nuclear/química , Tamanho da Partícula , Plasmídeos , Polietilenoimina/efeitos adversos , Polietilenoimina/síntese química , Ácido Poliglutâmico/efeitos adversos , Ácido Poliglutâmico/síntese química , Espectrometria de Fluorescência , Propriedades de Superfície , Transfecção
11.
Nanotechnology ; 22(37): 375102, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21852739

RESUMO

Non-viral gene delivery systems based on cationic polymers have faced limitations related to their relative low gene transfer efficiency, cytotoxicity and system instability in vivo. In this paper, a flexible and pompon-like dendrimer composed of poly (amidoamine) (PAMAM) G4.0 as the inner core and poly (L-glutamic acid) grafted low-molecular-weight polyethylenimine (PLGE) as the surrounding multiple arms was synthesized (MGI dendrimer). The novel MGI dendrimer was designed to combine the merits of size-controlled PAMAM G4.0 and the low toxicity and flexible chains of PLGE. In phosphate-buffered saline dispersions the well-defined DNA/MGI complex above a N/P ratio of 30 showed good stability with particle sizes of approximately 200 nm and a comparatively low polydispersity index. However, the particle size of the DNA/25 kDa polyethylenimine (DNA/PEI 25K) complex was larger than 700 nm under the same salt conditions. The shielding of the compact amino groups at the periphery of flexible PAMAM and biocompatible PLGE chains in MGI resulted in a dramatic decrease of the cytotoxicity compared to native PAMAM G4.0 dendrimer. The in vitro transfection efficiency of DNA/MGI dendrimer complex was higher than that of PAMAM G4.0 dendrimer. Importantly, in serum-containing medium, DNA/MGI complexes at their optimal N/P ratio maintained the same high levels of transfection efficiency as in serum-free medium, while the transfection efficiency of native PAMAM G4.0, PEI 25K and Lipofectamine 2000 were sharply decreased. In vivo gene delivery of pVEGF165/MGI complex into balloon-injured rabbit carotid arteries resulted in significant inhibition of restenosis by increasing VEGF165 expression in local vessels. Therefore, the pompon-like MGI dendrimer may be a promising vector candidate for efficient gene delivery in vivo.


Assuntos
Materiais Biocompatíveis/química , Dendrímeros/química , Técnicas de Transferência de Genes , Ácido Poliglutâmico/química , Animais , Células CHO , Sobrevivência Celular , Dicroísmo Circular , Cricetinae , Cricetulus , DNA/metabolismo , Desoxirribonucleases/metabolismo , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Espectroscopia de Ressonância Magnética , Peso Molecular , Neointima/patologia , Tamanho da Partícula , Plasmídeos/genética , Polietilenoimina/química , Coelhos , Espectrofotometria Infravermelho , Transfecção
12.
Ann Palliat Med ; 10(8): 9259-9266, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34488411

RESUMO

BACKGROUND: Hypertensive disorders of pregnancy (HDP) is a disease associated with elevated blood pressure during pregnancy, accounting for 5-10% of all pregnancies, which includes: gestational hypertension, preeclampsia (PE), eclampsia, chronic hypertension with superimposed PE and chronic hypertension. PE is the most prevalent type of HDP that seriously threatens the life and health of mothers and infants. In-depth exploration of the pathogenesis can play an early role in predicting the disease. METHODS: A literature search was conducted in PubMed, Embase, Google Scholar, and other databases in the article. It was investigated by searching for literature published between 1993 and March 2021; the subject terms included-"vascular endothelial growth factor", "preeclampsia", and "pathogenesis". In the article, the inclusion criteria of literature should meet the definition of PE. It was excluded as reviews, case reports, narrative reviews, and publications that lack key information. RESULTS: Vascular endothelial growth factor (VEGF) family factor research provides pivotal value for early clinical prediction of PE. Soluble fms-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PlGF) became a marker for early prediction of PE. Through the included 51 articles, the analysis of VEGF in PE and its pathway factors was summarized to clarify the pathogenesis further and provide innovative ideas for future research directions and clinical diagnosis. DISCUSSION: A systematic review of the VEGF family in the pathogenesis of PE was concluded in the study to find angiogenesis markers of PE from the pathogenesis of the available literature. Therefore, early intervention of clinical diseases could reduce maternal complications and ensure the maximum health of mothers and babies. There are differences in the research results of factors in the VEGF family, and further research is needed to provide accurate clinical evidence.


Assuntos
Pré-Eclâmpsia , Fator A de Crescimento do Endotélio Vascular , Biomarcadores , Feminino , Humanos , Fator de Crescimento Placentário , Gravidez , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
13.
Front Hum Neurosci ; 14: 360, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33192381

RESUMO

Sleep deprivation (SD) is very common in modern society and has a profound effect on cognitive function, in particular on working memory (WM). This type of memory is required for completion of many tasks and is adversely affected by SD. However, the cognitive neural mechanism by which SD affects WM, remains unclear. In this study, we investigated the changes in the brain network involved in WM after SD. Twenty-two healthy subjects underwent functional magnetic resonance imaging scan while in a state of resting wakefulness and again after 36 h of total SD and performed a WM task before each scanning session. Nineteen main nodes of the default mode network (DMN), dorsal attention network (DAN), fronto-parietal network (FPN), salience network (SN), and other networks were selected for functional analysis of brain network connections. Functional connectivity measures were computed between seed areas for region of interest (ROI)-to-ROI analysis and to identify patterns of ROI-to-ROI connectivity. The relationship between the significant changes in functional connectivity in the brain network and WM performance were then examined by Pearson's correlation analysis. WM performance declined significantly after SD. Compared with the awake state, the functional connectivity between DAN and DMN significantly increased after SD while that between FPN and DMN significantly decreased. Correlation analysis showed that the enhanced functional connectivity between DAN and DMN was negatively correlated with the decline in WM performance and that the decline in functional connectivity between FPN and DMN was positively correlated with decreased WM performance. These findings suggested that SD may affect WM by altering the functional connectivity among DMN, DAN, and FPN.

14.
Theranostics ; 10(15): 6581-6598, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32550891

RESUMO

Background: Exosome (Exo)-based chemotherapeutic drug delivery systems have been extensively investigated; however, the therapeutic potential of other subtypes of extracellular vesicles (EVs), in particular microvesicles (MiV), seem to be overlooked. Moreover, despite a general agreement on organ tropism of EVs, few studies have clearly demonstrated that EVs specifically target tumor tissue. Methods: Proinflammatory macrophage-derived EV subpopulations comprising apoptotic bodies (ApB), MiV and Exo were isolated under differential ultracentrifugation, and further analyzed using comparative proteomic and lipid approach. Results: On the basis of EV biogenesis pathways, our data demonstrated that MiV acquire the tumor-targeting capacity probably through inheritance of CCR2-enriched cell membrane which also drives the recruitment of donor cells to tumor sites. Further, our data validate MiV utilize SNARE-mediated membrane fusion to directly discharge doxorubicin to nucleus and bypass endocytic degradation. Conclusions: Compared with other EV subtypes, MiV loaded with doxorubicin gain significant benefits in chemotherapeutic outcomes including survival rate improvements in metastatic ovarian cancer. Therefore, MiV represent a potent alterative to Exo and synthetic liposomes (Lipo) for tumor-targeting drug delivery.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Quimiocina CCL2/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Macrófagos/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Receptores CCR2/metabolismo , Proteínas SNARE/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Doxorrubicina/farmacologia , Feminino , Humanos , Macrófagos/imunologia , Fusão de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proteômica/métodos , Receptores CCR2/genética , Proteínas SNARE/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biomater Sci ; 8(4): 1117-1126, 2020 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-31724666

RESUMO

Surgical resection currently remains the mainstay of treatment for patients with gliomas of any grade. The maximum extent of surgical resection is associated with a long-term disease control; however, maximal resection of the brain tumor possibly results in additional neurological deficits. Therefore, improving the precision in brain tumor surgery by visual identification and screening of tumor cells can help to tackle this devastating disease. In the present study, BV2 microglial cells were engineered by iron oxide-nanoparticle stimulation as intraoperative optical imaging agent vehicles and loaded with near-infrared fluorescent dye DiD (DiDBV2-Fe) potentially for fluorescence-guided brain tumor surgery. Activation of BV2 microglial cells by citrate-stabilized iron oxide nanoparticles at a concentration of 62.5 µg mL-1 significantly inhibited M2 markers (arginase-1 and CD206), which is able to minimize risks of the immunosuppressive effects caused by the M2-like phenotype of microglial cells. Meanwhile, activated BV2 microglial cells showed up-regulation of arylsulfatase A, apolipoprotein E, transferrin, and ferritin heavy chain-1 gene expression that tends to promote microglia transport across the blood-brain barrier (BBB). Compared to DiDBV2 without iron oxide activation, DiDBV2-Fe indicated strong tumor tropism in response to monocyte chemoattractant protein-1 (CCL2) secreted by U87MG tumor cells. In vivo experiments proved that DiDBV2-Fe efficiently crossed the BBB and more than 90% fluorescence intensity generated by activated microglial cells was detected in the brain when administered through the carotid artery in an orthotopic glioblastoma mouse model. Notably, DiDBV2-Fe produced clear tumor border demarcation on near-infrared imaging and exhibited a superior tumor-to-brain fluorescence ratio to commercial 5-aminolevulinic acid. Accumulated DiDBV2-Fe induced a strong fluorescence signal in brain tumor tissue for a prolonged period (4-24 h), which is beneficial to perform complex and time-consuming brain operations. Overall, our study suggests that this newly engineered microglial cell has promise for enabling more accurate brain tumor imaging for fluorescence-guided resections.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Engenharia Celular/métodos , Corantes Fluorescentes/administração & dosagem , Glioma/diagnóstico por imagem , Microglia/citologia , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular , Corantes Fluorescentes/química , Glioma/metabolismo , Humanos , Nanopartículas de Magnetita , Camundongos , Microglia/química , Microglia/metabolismo , Transplante de Neoplasias , Imagem Óptica , Ratos
16.
Theranostics ; 9(23): 6936-6948, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31660078

RESUMO

Objective: Engineered immune cells (e.g., therapeutic T cells) provide a revolutionary approach to combat cancer. Certain activated immune cells can exquisitely sense and respond to the tumor microenvironment. Here, we propose a paradigm based on engineering macrophages to allow selective intercellular drug delivery and augmentation of antitumor activities by hijacking tumor microtube networks. Methods: Macrophages were engineered via anchoring lipopolysaccharides on the plasma membrane (LM). The tumor tropism of LM encapsulating doxorubicin (LM-Dox) was monitored by a real-time cell migration assay and small animal in vivo imaging. Monocyte chemoattractant protein-1 (CCL2) was measured by quantitative PCR and ELISA. Intercellular conduit formation was characterized by confocal laser scanning microscopy and scanning electron microscopy. LM-Dox activation of tumor-associated macrophages to release TNF-α was evaluated by western blot and immunofluorescence assays. The potential therapeutic effects of LM-Dox in a 3D tumor-immune model and a murine orthotopic lung cancer model were tested. Results: LM-Dox exhibited tumor tropism in response to CCL2 produced by A549 lung tumor cells and lung tumor tissues resulting in a remarkably higher amount of tumor accumulation than the case of Lipo-Dox (~ 4-fold). Intriguingly, LM-Dox accumulated at tumor sites hijacked the established tumor microtube networks and even stimulated microtube formation with tumor cells but not with normal cells to enable selective and rapid transport of the drug to tumor cells. Simultaneously, LM-Dox induced secretion of TNF-α in tumor-associated macrophages, which increased the antitumor activity of Dox. Thus, LM-Dox increased the inhibitory effects on tumor growth and metastasis in a mouse orthotopic lung cancer model and minimized the side effects of Dox-induced tumor invasion. Conclusion: Lipopolysaccharide-anchored macrophages that can hijack tumor microtube networks for selective drug transport may serve as versatile bioactive carriers of anticancer drugs. In the clinical context, these engineered microphages represent a personalized medicine approach that can be translated into potential use of patient-derived monocytes/macrophages for drug delivery by means of cell-to-cell communication.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/química , Sistemas de Liberação de Medicamentos/métodos , Lipopolissacarídeos/química , Neoplasias Pulmonares/tratamento farmacológico , Macrófagos/química , Células A549 , Animais , Membrana Celular/química , Movimento Celular/efeitos dos fármacos , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Sistemas de Liberação de Medicamentos/instrumentação , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/fisiopatologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
ACS Nano ; 13(2): 1078-1096, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30608136

RESUMO

It is extremely difficult for cancer chemotherapy to control the peritoneal metastasis of advanced ovarian carcinoma given its inability to target disseminated tumors and the severe toxic side effects on healthy organs. Here, we report antitumor M1 macrophages developed as live-cell carriers that deliver anticancer drugs for the treatment of the metastatic ovarian carcinoma. Engineered doxorubicin-loaded M1 macrophages (M1-Dox) significantly enhanced tumor tropism by upregulation of CCR2 and CCR4 compared with their parent cells. Meanwhile, M1-Dox inhibited doxorubicin-induced tumor invasion, whereas commercial Lipo-Dox did not limit these side effects. Importantly, our data uncovered a drug delivery mechanism by which M1-Dox transferred drug cargoes into tumor cells  via a tunneling nanotube pathway. The tunneling nanotube network acted as a transportation expressway for ultrafast drug delivery of M1-Dox, leading to efficient ovarian carcinoma cell death. Furthermore, genetic, pharmacological, and physical perturbations of these tunneling nanotubes obviously decreased drug transfer of M1-Dox, which further validated the evident correlation between drug delivery of M1-Dox and tunneling nanotubes. Finally, in peritoneal metastatic ovarian carcinoma-burdened mice, M1-Dox specifically penetrated into and accumulated deep within disseminated neoplastic lesions compared with commercial Lipo-Dox, resulting in reducing metastatic tumors to a nearly undetectable level and significantly increasing overall survival. Overall, the strategy of engineered macrophages for ultrafast and accurate drug delivery via the tunneling nanotubular expressway potentially revolutionizes the treatment of metastatic ovarian carcinoma.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Macrófagos/química , Nanopartículas/química , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Macrófagos/metabolismo , Camundongos , Nanopartículas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/secundário , Tamanho da Partícula , Células RAW 264.7 , Propriedades de Superfície , Células Tumorais Cultivadas
18.
Adv Sci (Weinh) ; 6(5): 1802057, 2019 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-30886812

RESUMO

Advanced melanoma can rarely be cured. Photodynamic therapy (PDT) readily eradicates the primary melanoma but has limited ability to destroy the spreading tumor cells unless supported by other combinative interventions to augment systemic antitumor immunity. Based on the previously synthesized penetration-enhancing biomaterials, a topically administered nanoformulation is developed, which profoundly assists 5-aminolevulinic acid (5-ALA) in circumventing skin barrier to be selectively delivered to tumor cells. After endocytosis, accumulated 5-ALA is efficiently metabolized to a photosensitizer protoporphyrin IX (PpIX) which stimulates a large production of cytotoxic reactive oxygen species (ROS) under illumination. Accompanied by the robust inflammatory responses followed by primary tumor destruction, CD4+CD8+ double positive T cells are highly boosted to harness host immunity to purge metastases in lymphoid organs. Compared with dacarbazine and programmed death 1 (PD-1) antibody, this treatment in advanced melanoma murine models, achieves a striking curable rate of 90% without melanoma prognostic markers LDH and S-100B detection, followed by a relapse-free survival rate of 83.33% in 300 days. Moreover, the cured mice's immune system function recovers to an extent similar to healthy mice without prolonged or exaggerated inflammation. This study using the synergistic biomaterials approach may thus render 5-ALA-mediated PDT a potentially curative therapy for advanced melanoma in clinic.

19.
Biomaterials ; 183: 77-92, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30149232

RESUMO

Many human cancer cells exhibit an oncogenetic-driven addiction to glutamine (Gln) as rapidly proliferating cancer cells consume Gln at a dramatically increased rate compared to normal cells. Tumor cells, therefore, compete with host cells for Gln, which causes Gln to flux from normal tissues to the tumor. We have developed and characterized a Gln macromolecular analog polyglutamine (PGS) for the delivery of gene regulators, such as siRNAs, in our previous works. Here, we hypothesize that PGS can utilize the Gln transporter SLC1A5 to specifically deliver therapeutic compounds to Gln-addicted cancer cells. Compared to human lung fibroblast HLF cells, cisplatin-resistant human lung adenocarcinoma A549/DDP cells significantly overexpress SLC1A5, which has a high binding affinity to PGS, as confirmed through molecular docking analysis. Due to the differences in Gln metabolism between malignant and normal cells, PGS/siRNA complexes were remarkably increased in cancer cells, especially when cells were deprived of Gln, which mirrors the conditions that are commonly found in a tumor microenvironment. Furthermore, we identified that chemical and genetic inhibition of Gln transporter SLC1A5 reduced the cellular internalization of PGS/siRNA complexes, suggesting a critical role for SLC1A5 in PGS uptake in cells. In turn, PGS upregulated SLC1A5 expression. Increased uptake of PGS complexes profoundly decreased intracellular Gln levels. Decreased Gln caused a moderate reduction in cell growth. To restore drug sensitivity and further enhance anti-tumor effects, the hybrid siRNAs anti-Survivin and anti-MDR1 (siSM), as model therapeutics, were administered through the PGS delivery system, which resulted in knockdown of Survivin and MDR1 and further sensitized cancer cells to the drug cisplatin (DDP). Since PGS complexes administered i.v. mostly accumulated in the lung parenchyma, a lung orthotopic tumor model was established to evaluate their inhibitory effects on tumors in the lungs. PGS/siSM comparably decreased the rate of tumor growth, while concurrent administration of PGS/siSM and DDP enhanced this effect and insignificantly improved life span. Consistent with our hypothesis, this study demonstrated that PGS mimicked Gln in the SLC1A5 pathway and selectively ferried therapeutics to Gln-addicted cancer cells. Our findings identified a new lung cancer targeting strategy based on Gln metabolism and can be used as a drug/gene delivery system.


Assuntos
Sistema ASC de Transporte de Aminoácidos/metabolismo , Portadores de Fármacos/química , Glutamina/metabolismo , Neoplasias Pulmonares/terapia , Antígenos de Histocompatibilidade Menor/metabolismo , Nanopartículas/química , Peptídeos/química , RNA Interferente Pequeno/administração & dosagem , Sistema ASC de Transporte de Aminoácidos/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Cisplatino/farmacologia , Liberação Controlada de Fármacos , Resistencia a Medicamentos Antineoplásicos , Terapia Genética , Humanos , Neoplasias Pulmonares/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Antígenos de Histocompatibilidade Menor/genética , RNA Interferente Pequeno/genética , Microambiente Tumoral
20.
J Control Release ; 262: 305-316, 2017 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-28797579

RESUMO

Drug nanocarriers that have long been expected to revolutionize cancer therapy have yet to achieve the significant clinical success. Therefore, it remains controversial to pursue a complex drug nanocarrier that lacks clinical relevance. Herein, we developed an easily-synthesized anti-tumor drug that actively seeks the acidic tumor microenvironment while ignoring the normal tissue without the aid of additional carriers. This called "carrier-free" drug (CFD) is capable of switching its morphology from the unstructured solution to the spherical structure in response to tumor acidity. CFDs were the water-soluble zwitterionic unimers to prevent the non-specific distribution in the circulation, whereas they spontaneously formed into the particles tending to accumulation in tumor. CFD overloading in tumor cells caused the lysosomal dysfunction and autophagy blockage, thereby triggered the cell death. All the in vitro and in vivo data demonstrated the tumor-acidity-selective cytotoxicity of CFD. This facile strategy to create a self-delivering anticancer drug may cast a new light on the development of cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/química , Microambiente Tumoral , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Soluções , Carga Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA