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












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 454, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212623

RESUMEN

Emerging evidence indicates that the activation of ferroptosis by glutathione peroxidase 4 (GPX4) inhibitors may be a prominent therapeutic strategy for tumor suppression. However, the wide application of GPX4 inhibitors in tumor therapy is hampered due to poor tumor delivery efficacy and the nonspecific activation of ferroptosis. Taking advantage of in vivo self-assembly, we develop a peptide-ferriporphyrin conjugate with tumor microenvironment specific activation to improve tumor penetration, endocytosis and GPX4 inhibition, ultimately enhancing its anticancer activity via ferroptosis. Briefly, a GPX4 inhibitory peptide is conjugated with an assembled peptide linker decorated with a pH-sensitive moiety and ferriporphyrin to produce the peptide-ferriporphyrin conjugate (Gi-F-CAA). Under the acidic microenvironment of the tumor, the Gi-F-CAA self-assembles into large nanoparticles (Gi-F) due to enhanced hydrophobic interaction after hydrolysis of CAA, improving tumor endocytosis efficiency. Importantly, Gi-F exhibits substantial inhibition of GPX4 activity by assembly enhanced binding (AEB) effect, augmenting the oxidative stress of ferriporphyrin-based Fenton reaction, ultimately enabling antitumor properties in multiple tumor models. Our findings suggest that this peptide-ferriporphyrin conjugate design with AEB effect can improve the therapeutic effect via induction of ferroptosis, providing an alternative strategy for overcoming chemoresistance.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Endocitosis , Hemina , Hidrólisis , Péptidos/farmacología , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
2.
Angew Chem Int Ed Engl ; 62(37): e202308049, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37486792

RESUMEN

Proteolysis targeting chimera (PROTAC) is an emerging pharmacological modality with innovated post-translational protein degradation capabilities. However, off-target induced unintended tissue effects and intrinsic "hook effect" hinder PROTAC biotechnology to be maturely developed. Herein, an intracellular fabricated nano proteolysis targeting chimeras (Nano-PROTACs) modality with a center-spoke degradation network for achieving efficient dose-dependent protein degradation in tumor is reported. The PROTAC precursors are triggered by higher GSH concentrations inside tumor cells, which subsequently in situ self-assemble into Nano-PROTACs through intermolecular hydrogen bond interactions. The fibrous Nano-PROTACs can form effective polynary complexes and E3 ligases degradation network with multi-binding sites, achieving dose-dependent protein degradation with "anti-hook effect". The generality and efficacy of Nano-PROTACs are validated by degrading variable protein of interest (POI) such as epidermal growth factor receptor (EGFR) and androgen receptor (AR) in a wide-range dose-dependent manner with a 95 % degradation rate and long-lasting potency up to 72 h in vitro. Significantly, Nano-PROTACs achieve in vivo dose-dependent protein degradation up to 79 % and tumor growth inhibition in A549 and LNCap xenograft mice models, respectively. Taking advantages of in situ self-assembly strategy, the Nano-PROTACs provide a generalizable platform to promote precise clinical translational application of PROTAC.


Asunto(s)
Neoplasias , Ubiquitina-Proteína Ligasas , Humanos , Animales , Ratones , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas/metabolismo , Sitios de Unión
3.
Adv Mater ; 35(45): e2303831, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37462447

RESUMEN

Anti-PD-L1 monoclonal antibody has achieved substantial success in tumor immunotherapy by T-cells activation. However, the excessive accumulation of extracellular matrix components induced by unsatisfactory T-cells infiltration and poor tumor penetration of antibodies make it challenging to realize efficient tumor immunotherapy. Herein, a peptide-based bispecific nanoblocker (BNB) strategy is reported for in situ construction of CXCR4/PD-L1 targeted nanoclusters on the surface of tumor cells that are capable of boosting T-cells infiltration through CXCR4 blockage and enhancing T-cells activation by PD-L1 occupancy, ultimately realizing high-performance tumor immunotherapy. Briefly, the BNB strategy selectively recognizes and bonds CXCR4/PD-L1 with deep tumor penetration, which rapidly self-assembles into nanoclusters on the surface of tumor cells. Compared to the traditional bispecific antibody, BNB exhibits an intriguing metabolic behavior, that is, the elimination half-life (t1/2 ) of BNB in the tumor is 69.3 h which is ≈50 times longer than that in the plasma (1.4 h). The higher tumor accumulation and rapid systemic clearance overcome potential systemic side effects. Moreover, the solid tumor stress generated by excessive extracellular matrix components is substantially reduced to 44%, which promotes T-cells infiltration and activation for immunotherapy efficacy. Finally, these findings substantially strengthen and extend clinical applications of PD-1/PD-L1 immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Humanos , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Neoplasias/terapia , Anticuerpos Biespecíficos/uso terapéutico , Linfocitos T/metabolismo , Inmunoterapia
4.
Cell Prolif ; 56(12): e13494, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37139662

RESUMEN

Acute liver injury (ALI) is a severe liver disease that is characterized by sudden and massive hepatocyte necrosis and deterioration of liver functions. Oxidative stress is increasingly recognized as a key factor in the induction and progression of ALI. Scavenging excessive reactive oxygen species (ROS) with antioxidants has become a promising therapeutic option, but intrinsically hepatocyte-targeting antioxidants with excellent bioavailability and biocompatibility are yet to be developed. Herein, self-assembling nanoparticles (NPs) composed of amphiphilic polymers are introduced to encapsulate organic Selenium compound L-Se-methylselenocysteine (SeMC) and form SeMC NPs, which protect the viabilities and functions of cultured hepatocytes in drug- or chemical-induced acute hepatotoxicity models via efficient ROS removal. After further functionalization with the hepatocyte-targeting ligand glycyrrhetinic acid (GA), the resultant GA-SeMC NPs exhibit enhanced hepatocyte uptake and liver accumulation. In mouse models of ALI induced by acetaminophen (APAP) or carbon tetrachloride (CCl4 ), treatment with GA-SeMC NPs significantly decrease the levels of hepatic lipid peroxidation, tissue vacuolization and serum liver transaminases, while prominently increase that of endogenous antioxidant enzymes. Our study therefore presents a liver-targeting drug delivery strategy for the prevention and treatment of hepatic diseases.


Asunto(s)
Antioxidantes , Ácido Glicirretínico , Ratones , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Hígado/metabolismo , Hepatocitos , Estrés Oxidativo , Ácido Glicirretínico/metabolismo , Ratones Endogámicos C57BL
5.
Adv Mater ; 35(24): e2210458, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37046183

RESUMEN

Germline stem cells (GSCs) are the only cell population capable of passing genetic information to offspring, making them attractive targets in reproductive biology and fertility research. However, it is generally more difficult to introduce exogenous biomolecules into GSCs than other cell types, impeding the exploration and manipulation of these cells for biomedical purposes. Herein, semiconductor polymer dots (Pdots)-based nanocomplex Pdot-siRNA is developed and achieves effective knockdown of target genes in female germline stem cells (FGSCs). Advantage of high fluorescence brightness of Pdots is taken for comprehensive investigation of their cellular uptake, intracellular trafficking, and exocytosis in FGSCs. Importantly, Pdots show excellent biocompatibility and minimally disturb the differentiation of FGSCs. Intracellular Pdots escape from the lysosomes and undergo active exocytosis, which makes them ideal nanocarriers for bioactive cargos. Moreover, Pdot-siRNA can penetrate into 3D ovarian organoids derived from FGSCs and down-regulate the expression levels of target genes. This study investigates the interface between a type of theranostic nanoparticles and FGSCs for the first time and sheds light on the manipulation and medical application of FGSCs.


Asunto(s)
Nanopartículas , Células Madre Oogoniales , Puntos Cuánticos , Polímeros , Semiconductores , Exocitosis
6.
Sci Adv ; 9(9): eabq8225, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36857458

RESUMEN

Up to 75% of bladder cancer patients suffer from recurrence due to postoperative tumor implantation. However, clinically used Bacillus Calmette-Guerin (BCG) treatment failed to inhibit the recurrence. Here, we report a bispecific glycopeptide (bsGP) that simultaneously targets CD206 on tumor-associated macrophages (TAMs) and CXCR4 on tumor cells. bsGP repolarizes protumoral M2-like TAMs to antitumor M1-like that mediated cytotoxicity and T cell recruitment. Meanwhile, bsGP is cleaved by the MMP-2 enzyme to form nanostructure for the long-term inhibition of CXCR4 downstream signaling, resulting in reduced tumor metastasis and promoted T cell infiltration. In orthotopic bladder tumor models, bsGP reduced the postoperative recurrence rate to 22%. In parallel, the recurrence rates of 89 and 78% were treated by doxycycline and BCG used in clinic, respectively. Mechanistic studies reveal that bsGP reduces the matrix microenvironment barrier, increasing the spatially redirected CD8+ T cells to tumor cells. We envision that bis-targeting CD206 and CXCR4 may pave the way to inhibit tumor metastasis and recurrence.


Asunto(s)
Microambiente Tumoral , Neoplasias de la Vejiga Urinaria , Humanos , Vacuna BCG , Linfocitos T CD8-positivos , Recurrencia Local de Neoplasia , Glicopéptidos
7.
ACS Appl Mater Interfaces ; 15(3): 3839-3850, 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36637993

RESUMEN

Framework nucleic acids (FNAs) represent a new type of DNA-based nanomaterials and possess great potentials in biosensing, bioimaging, and molecular delivery. Hierarchical DNA nanostructures that consist of multiple FNA monomers increase the capacity for drug delivery and multifunctional modification. However, there are relatively few studies devoted to the behavior and regulation of hierarchical FNAs in living cells, impeding their further applications. Herein, we constructed a dendritic nanostructure with five tetrahedral DNA nanocages and characterized the real-time internalization, inter-organelle trafficking, and exocytosis in living mammalian cells. In comparison to FNA monomers, FNA dendrimers exhibit increased endocytosis and prolonged cellular retention. Single-particle tracking on hundreds of FNA dendrimers exhibits no interference on the mobility or kinetics of subcellular organelles, implying that FNAs as well as their higher-order derivatives are ideal intracellular imaging probes and nanocarriers. Our study validates the suitability and superiority of hierarchical DNA nanostructures as high-valency scaffolds for biomedical applications.


Asunto(s)
Dendrímeros , Nanoestructuras , Ácidos Nucleicos , Animales , Ácidos Nucleicos/química , Dendrímeros/química , ADN/química , Nanoestructuras/química , Diagnóstico por Imagen , Mamíferos
8.
Biomaterials ; 287: 121655, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35810541

RESUMEN

Aggregation-induced emission luminogens (AIEgens) possess enhanced fluorescence in highly aggregated states, thus enabling AIEgens as a promising module for highly emissive fluorescence biomaterials. So far, AIEgens-based nanomaterials and their hybrids have been reported for biomedical applications. Benefiting from the intrinsic biocompatibility and biofunction-editing properties of peptides, peptide-AIEgens hybrid biomaterials reveal unlimited possibilities including target capacity, specificity, stimuli-responsiveness, self-assembly, controllable structural transformation, etc.. In the last two decades, peptide-AIEgens hybrid nanomaterials with a unique design concept in aggregated states have achieved various biomedical applications such as biosensing, bioimaging, imaging-guided surgery, drug delivery and therapy. More recently, programmable design of peptide-AIEgens for in situ self-assembly provides a unique strategy for constructing intelligent entities with defined biological functions. In this review, we summarize the basic design principle of programmable peptide-AIEgens, structure-effect relationship and their unusual biomedical effects. Finally, an outlook and perspective toward future challenges and developments of peptide-AIEgens nanomaterials are concluded.

9.
Nano Lett ; 22(10): 3983-3992, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35548949

RESUMEN

Lysosome-targeting self-assembling prodrugs had emerged as an attractive approach to overcome the acquisition of resistance to chemotherapeutics by inhibiting lysosomal sequestration. Taking advantage of lysosomal acidification induced intracellular hydrolytic condensation, we developed a lysosomal-targeting self-condensation prodrug-nanoplatform (LTSPN) system for overcoming lysosome-mediated drug resistance. Briefly, the designed hydroxycamptothecine (HCPT)-silane conjugates self-assembled into silane-based nanoparticles, which were taken up into lysosomes by tumor cells. Subsequently, the integrity of the lysosomal membrane was destructed because of the acid-triggered release of alcohol, wherein the nanoparticles self-condensed into silicon particles outside the lysosome through intracellular hydrolytic condensation. Significantly, the LTSPN system reduced the half-maximal inhibitory concentration (IC50) of HCPT by approximately 4 times. Furthermore, the LTSPN system realized improved control of large established tumors and reduced regrowth of residual tumors in several drug-resistant tumor models. Our findings suggested that target destructing the integrity of the lysosomal membrane may improve the therapeutic effects of chemotherapeutics, providing a potent treatment strategy for malignancies.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Línea Celular Tumoral , Resistencia a Medicamentos , Humanos , Lisosomas/patología , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Profármacos/farmacología , Profármacos/uso terapéutico , Silanos/farmacología , Silanos/uso terapéutico
10.
Bioact Mater ; 14: 110-119, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35310363

RESUMEN

Intraoperative fluorescence-based tumor imaging plays a crucial role in performing the oncological safe tumor resection with the advantage of differentiating tumor from normal tissues. However, the application of these fluorescence contrast agents in renal cell carcinoma (RCC) and hepatocellular carcinoma (HCC) was dramatically hammered as a result of lacking active targeting and poor retention time in tumor, which limited the Signal to Noise Ratio (SNR) and narrowed the imaging window for complicated surgery. Herein, we reported an activated excretion-retarded tumor imaging (AERTI) strategy, which could be in situ activated with MMP-2 and self-assembled on the surface of tumor cells, thereby resulting in a promoted excretion-retarded effect with an extended tumor retention time and enhanced SNR. Briefly, the AERTI strategy could selectively recognize the Integrin αvß3. Afterwards, the AERTI strategy would be activated and in situ assembled into nanofibrillar structure after specifically cleaved by MMP-2 upregulated in a variety of human tumors. We demonstrated that the AERTI strategy was successfully accumulated at the tumor sites in the 786-O and HepG2 xenograft models. More importantly, the modified modular design strategy obviously enhanced the SNR of AERTI strategy in the imaging of orthotopic RCC and HCC. Taken together, the results presented here undoubtedly confirmed the design and advantage of this AERTI strategy for the imaging of tumors in metabolic organs.

11.
Angew Chem Int Ed Engl ; 61(18): e202116893, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35181975

RESUMEN

Intravesical administration of first-line drugs has shown failure in the treatment of bladder cancer owing to the poor tumor retention time of chemotherapeutics. Herein, we report an intracellular hydrolytic condensation (IHC) system to construct long-term retentive nano-drug depots in situ, wherein sustained drug release results in highly efficient suppression of bladder cancer. Briefly, the designed doxorubicin (Dox)-silane conjugates self-assemble into silane-based prodrug nanoparticles, which condense into silicon particle-based nano-drug depots inside tumor cells. Significantly, we demonstrate that the IHC system possesses highly potent antitumor efficacy, which leads to the regression and eradication of large established tumors and simultaneously extends the overall survival of air pouch bladder cancer mice compared with that of mice treated with Dox. The concept of intracellular hydrolytic condensation can be extended via conjugating other chemotherapeutic drugs, which may facilitate rational design of novel nanomedicines for augmentation of chemotherapy.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias de la Vejiga Urinaria , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Portadores de Fármacos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Masculino , Ratones , Nanopartículas/uso terapéutico , Silanos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
12.
Biomaterials ; 278: 121139, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34624753

RESUMEN

Peptide drug conjugate (PDC) has emerged as one of the new generations of targeted therapeutics for cancer, which owns the advantages of improved drug targetability and reduced adverse effects compared with traditional chemotherapy. However, the poor permeability of PDC drugs regarding tumor cells is an urgent problem to be solved. Herein, we design a PDC drug molecule, which is composed of three modules: targeting motif (RGD target), assembly motif (GNNNQNY) and cytotoxic payload (CPT molecule). This PDC in situ forms nanoclusters upon binding cellular receptor, resulting in improved PDC cell-entry efficiency and treatment efficacy. In addition, the PDC shows increased therapeutic efficacy and raises the maximum tolerance dose of the drug in breast and bladder xenografted mice models. This strategy leverages the assembly principle to promote penetration of peptide molecules into cells and increase intracellular drug bioavailability, which is of great significance for the development of PDC drugs in the future.


Asunto(s)
Antineoplásicos , Preparaciones Farmacéuticas , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ratones , Péptidos
13.
Nano Lett ; 20(7): 5228-5235, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32510963

RESUMEN

Cell-membrane-camouflaged nanoparticles (CMC-NPs) have been increasingly exploited to develop various therapeutic tools due to their high biocompatibility and cell-type-specific tumor-targeting properties. However, the molecular mechanism of CMC-NPs for homotypic targeting remains elusive. Here, we develop a plasmonic imaging method by coating gold nanoparticles (AuNPs) with cancer cell membranes and perform plasmonic imaging of the interactions between CMC-NPs and living cells at the single-cell level. Quantitative analysis of CMC-NPs in a different clustering status reveals that the presence of cell membranes on CMC-NPs results in a 7-fold increase in homotypic cell delivery and nearly 2 orders of magnitude acceleration of the intracellular agglomeration process. Significantly, we identify that integrin αvß3, a cell surface receptor abundantly expressed in tumor cells, is critical for the selective cell recognition of CMC-NPs. We thus establish a single-cell plasmonic imaging platform for probing NP-cell interactions, which sheds new light on the therapeutic applications of CMC-NPs.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Línea Celular Tumoral , Membrana Celular , Oro , Integrina alfaVbeta3
14.
Cell Prolif ; 52(5): e12656, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31264309

RESUMEN

OBJECTIVES: Cell migration has a key role in cancer metastasis, which contributes to drug resistance and tumour recurrence. Better understanding of the mechanisms involved in this process will potentially reveal new drug targets for cancer therapy. Fer is a non-receptor protein tyrosine kinase aberrantly expressed in various human cancers, whereas its role in tumour progression remains elusive. MATERIALS AND METHODS: Transgenic flies and epigenetic analysis were employed to investigate the role of Drosophila Fer (FER) in cell migration and underlying mechanisms. Co-immunoprecipitation assay was used to monitor the interaction between FER and Drosophila JNK (Bsk). The conservation of Fer in regulating JNK signalling was explored in mammalian cancer and non-cancer cells. RESULTS: Overexpression of FER triggered cell migration and activated JNK signalling in the Drosophila wing disc. Upregulation and downregulation in the basal activity of Bsk exacerbated and eliminated FER-mediated migration, respectively. In addition, loss of FER blocked signal transduction of the JNK pathway. Specifically, FER interacted with and promoted the activity of Bsk, which required both the kinase domain and the C-terminal of Bsk. Lastly, Fer regulated JNK activities in mammalian cells. CONCLUSIONS: Our study reveals FER as a positive regulator of JNK-mediated cell migration and suggests its potential role as a therapeutic target for cancer metastasis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proteínas de Drosophila/química , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/química , Metaloproteinasa 1 de la Matriz/metabolismo , Dominios Proteicos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Alas de Animales/metabolismo
16.
Cell Prolif ; 52(2): e12556, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30710394

RESUMEN

OBJECTIVES: Nanocarriers can greatly enhance the cellular uptake of therapeutic agents to regulate cell proliferation and metabolism. Nevertheless, further application of nanocarriers is often limited by insufficient understanding of the mechanisms of their uptake and intracellular behaviour. MATERIALS AND METHODS: Fluorescent polymer dots (Pdots) are coated with synthetic octaarginine peptides (R8) and are analysed for cellular uptake and intracellular transportation in HeLa cervical cancer cells via single particle tracking. RESULTS: Surface modification with the R8 peptide efficiently improves both cellular uptake and endosomal escape of Pdots. With single particle tracking, we capture the dynamic process of internalization and intracellular trafficking of R8-Pdots, providing new insights into the mechanism of R8 in facilitating nanostructure-based cellular delivery. Furthermore, our results reveal R8-Pdots as a novel type of autophagy inducer. CONCLUSIONS: This study provides new insights into R8-mediated cellular uptake and endosomal escape of nanocarriers. It potentiates biological applications of Pdots in targeted cell imaging, drug delivery and gene regulation.


Asunto(s)
Endocitosis , Colorantes Fluorescentes/metabolismo , Oligopéptidos/metabolismo , Polímeros/metabolismo , Transporte Biológico , Endosomas/metabolismo , Colorantes Fluorescentes/análisis , Células HeLa , Humanos , Microscopía Confocal , Nanoestructuras/análisis , Oligopéptidos/análisis , Imagen Óptica , Polímeros/análisis
17.
Cell Prolif ; 52(1): e12529, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30328653

RESUMEN

OBJECTIVES: Over the past decade an intriguing connection between cell polarity and tumorigenesis has emerged. Multiple core components of the junction complexes that help to form and maintain cell polarity display both pro- and anti-tumorigenic functions in a context-dependent manner, with the underlying mechanisms poorly understood. MATERIALS AND METHODS: With transgenic fly lines that overexpress or knock down specific signalling components, we perform genetic analysis to investigate the precise role of the polarity protein Canoe (Cno) in tumorigenesis and the downstream pathways. RESULTS: We show that overexpression of cno simultaneously activates JNK and Ras-MEK-ERK signalling, resulting in mixed phenotypes of both overproliferation and cell death in the Drosophila wing disc. Moderate alleviation of JNK activation eliminates the effect of Cno on cell death, leading to organ overgrowth and cell migration that mimic the formation and invasion of tumours. In addition, we find that the Hippo pathway acts downstream of JNK and Ras signalling to mediate the effect of Cno on cell proliferation. CONCLUSIONS: Our work reveals an oncogenic role of Cno and creates a new type of Drosophila tumour model for cancer research.


Asunto(s)
Polaridad Celular/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas ras/metabolismo , Animales , Carcinogénesis/genética , Muerte Celular/genética , Proliferación Celular/genética , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/genética , Técnicas de Silenciamiento del Gen , Neoplasias/genética , Neoplasias/patología , Alas de Animales/embriología
18.
ACS Cent Sci ; 4(10): 1344-1351, 2018 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-30410972

RESUMEN

Cell entry of anionic nano-objects has been observed in various types of viruses and self-assembled DNA nanostructures. Nevertheless, the physical mechanism underlying the internalization of these anionic particles across the negatively charged cell membrane remains poorly understood. Here, we report the use of virus-mimicking designer DNA nanostructures with near-atomic resolution to program "like-charge attraction" at the interface of cytoplasmic membranes. Single-particle tracking shows that cellular internalization of tetrahedral DNA nanostructures (TDNs) depends primarily on the lipid-raft-mediated pathway, where caveolin plays a key role in providing the short-range attraction at the membrane interface. Both simulation and experimental data establish that TDNs approach the membrane primarily with their corners to minimize electrostatic repulsion, and that they induce uneven charge redistribution in the membrane under the short-distance confinement by caveolin. We expect that the nanoscale like-charge attraction mechanism provides new clues for viral entry and general rules for rational design of anionic carriers for therapeutics.

19.
Adv Mater ; 30(46): e1804861, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30276898

RESUMEN

Stem cells generally exist in low abundance and tend to lose stemness in the absence of self-renewal signals. While extracellular-matrix-mimicking techniques have been developed to support stem cell proliferation, the lack of niche cells in these synthetic systems often hampers continuous stem cell expansion and maintenance of pluripotency, which are indispensable for regenerative medicine. Here, an intercellular DNA-reaction-programmed ESPN (expansion of stem cells with pairing niches) strategy is developed for 3D culture of mammary stem cells (MaSCs). Boolean logic operations are implemented to confer DNA-programmed mechanical signaling and genetically engineered morphogen signaling by niche cells, resulting in sustained expansion of MaSCs in vitro. The creation of stem cell niches improves the proliferation of pluripotent cells by four times during one-week culture. This method thus provides a novel approach for logical regulation of stemness and proliferation of stem cells for biomedicine.


Asunto(s)
ADN/análisis , Ingeniería Genética/métodos , Células Madre Pluripotentes/citología , Células Madre/citología , Animales , Proliferación Celular , Separación Celular , Colágeno/química , Combinación de Medicamentos , Femenino , Citometría de Flujo , Células HEK293 , Humanos , Técnicas In Vitro , Laminina/química , Glándulas Mamarias Animales/citología , Ratones , Hibridación de Ácido Nucleico , Proteoglicanos/química , Transducción de Señal , Estrés Mecánico
20.
ACS Appl Mater Interfaces ; 10(4): 3449-3458, 2018 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-29318884

RESUMEN

Titanium dioxide nanoparticles (TiO2NPs) are among the most widely manufactured nanomaterials with broad applications in food industry, cosmetics, and medicine. Although the toxicity of TiO2NPs at high doses has been extensively explored, the potential health risks of TiO2NPs exposure at nontoxic concentrations remain poorly understood. Epithelial-mesenchymal transition (EMT) plays pivotal roles in a diversity of physiological and pathological processes, including tissue regeneration and cancer metastasis. In this study, we find that the cellular uptake of TiO2NPs inhibits EMT-mediated cell remodeling and cell migration without exhibiting cytotoxicity. Further investigation reveals that TiO2NPs suppress the process of EMT through the blockade of transforming growth factor-ß (TGFß) signaling. Particularly, TiO2NPs interact with the TGFß receptor TßRI/II complex, induce its lysosomal degradation, and thereby downregulate expression of TGFß target genes. Moreover, we show that waterborne TiO2NPs do not elicit toxicity in healthy tissues but hamper EMT-mediated wound healing in two animal models. Long-term exposure of TiO2NPs in environmental water and drinking water impede the regeneration of amputated fin in zebrafish and the recovery of intestinal mucosal damage in colitic mice. Our results reveal the previously unknown effects of TiO2NPs during tissue remodeling and repair, which have significant implications in their risk assessment and management.


Asunto(s)
Transición Epitelial-Mesenquimal , Animales , Ratones , Nanopartículas , Regeneración , Titanio , Factor de Crecimiento Transformador beta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...