Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Environ Toxicol ; 35(10): 1146-1156, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32511866

RESUMEN

Circular RNAs (CircRNAs) are a group of noncoding RNAs that have essential function in the development and progression of various cancers. The expression pattern and function of circRNA in lung cancer is not fully understood. In the present study, we aimed to investigate the expression profiles and underlying mechanism of circRNA circ_POLA2 in lung cancer cell stemness. Circ_POLA2 was highly expressed in lung cancer tissues and predicted a poor prognosis in lung cancer patients. Knockdown of circ_POLA2 inhibited the stemness of lung cancer cells, which is evident by the decreased sphere-formation ability, ALDH1 activity, and stemness marker expression, but had no effects on cell viability. Mechanistically, circ_POLA2 functioned as a ceRNA by sponging miR-326. Furthermore, miR-326 negatively regulated G protein subunit beta 1 (GNB1) expression by targeting its 3'-UTR (untranslated region). Intriguingly, we found that GNB1 was overexpressed and associated with poor prognosis in lung cancer patients. Overexpression of GNB1 could antagonize the inhibitory effect of circ_POLA2 knockdown on lung cancer cell stemness. In conclusion, circ_POLA2 promotes lung cancer cell stemness and progression via regulating the miR-326/GNB1 axis, which might serve as a novel therapeutic target for lung cancer patients.


Asunto(s)
ADN Polimerasa I/genética , Subunidades beta de la Proteína de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , MicroARNs/genética , Células Madre Neoplásicas/patología , ARN Circular/genética , Regiones no Traducidas 3'/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/patología
2.
Macromol Rapid Commun ; 40(8): e1800824, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30779386

RESUMEN

A tumor-selective drug delivery nanogel with redox-responsive size swelling and co-instantaneous drug release is developed. The nanogel is formed by poly(ethylene glycol) diglycidyl ether and cystamine double crosslinked hyaluronic acid (HA). The disulfide bond in cystamine (Cys) is in charge of the responsiveness, while the compact polymer network turns the nanogel a capsule for effective drug loading. The tumor targeting is achieved by the known HA-receptor mediated endocytosis. The responsive swelling of this nanogel and co-instantaneous drug releases happen with the cleavage of the disulfide bond following tumor targeting and cell endocytosis, which is triggered by massive glutathione (GSH) in the cytoplasm of tumor cells. The highly selective nanogel uptake by tumor cells is directly demonstrated by fluorescence microscopy and flow cytometry. The dynamic light scattering and fluorescent spectrum reveal the GSH-triggered size change and simultaneous drug release, which results in higher tumor cytotoxicity and over fourfold efficacy against tumor cells compared with normal cells. These results indicate that these HA-PEG-Cys-DOX nanogels, with performance of selective drug delivery, intracellular reconstruction, and responsive drug release, are promising platforms for better therapeutic effects in cancer treatment.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Neoplasias/tratamiento farmacológico , Polietilenglicoles/química , Polietileneimina/química , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular , Proliferación Celular/efectos de los fármacos , Cistamina/química , Doxorrubicina/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ácido Hialurónico/química , Nanogeles , Neoplasias/patología , Oxidación-Reducción
3.
Carbohydr Polym ; 313: 120868, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37182959

RESUMEN

Soft actuators and sensors have attracted extensive scientific interest attributed to their great potential applications in various fields, but the integration of actuating and sensing functions in one material is still a big challenge. Here, we developed an electrospun cellulose acetate (CA)/carbon nanotube nanofiborous composite with both functional applications as multi-responsive shape memory actuators and triboelectric nanogenerator (TENG) based sensors. Attributed to excellent thermo- and light-induced shape memory performance, the CA nanofiborous composites showed high heavy-lift capability as light driven actuators, able to lift burdens 1050 times heavier than their own weight. The CA nanofiborous membranes based TENG exhibited high output performance with open-circuit voltage, short-circuit density, and instantaneous power density about 103.2 V, 7.93 mA m-2 and 0.74 W m-2, respectively. The fabricated TENG based pressure sensor exhibited a high sensitivity of 3.03 V kPa-1 below 6.8 kPa and 0.11 V kPa-1 in the pressure range from 6.8 to 65 kPa, which can be effectively used to monitor human motion state and measure wind velocity. It is expected that the electrospun composites with actuating and sensing functions will show prosperous applications prospects in soft robotics.

4.
Am J Cancer Res ; 13(5): 2104-2115, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37293167

RESUMEN

Evidence suggests that immunogenic cell death (ICD) releases cancer antigens that promote cytotoxic T-cell responses, potentially improving immunotherapy. However, the relationship between ICDs and esophageal cancer (EC) remains unclear. This study aimed to determine the role of ICDs in EC and to construct an ICD-based prognostic panel. RNA-seq data of EC and the corresponding clinical information were downloaded from the UCSC-Xena platform to explore the association between ICD gene expression and EC prognosis. The GSE53625 dataset was used to validate the proposed model. Differentially expressed genes (DEGs) between different molecular subtypes were identified to construct a new ICD-related prognosis panel and generate molecular subtypes using ConsensusClusterPlus. We created a prognostic profile based on the ICD and a nomogram based on the risk score. Compared with normal samples, ICD gene expression of malignant samples were significantly increased. 161 patients with EC were successfully divided into three subtypes (SubA, SubB, and SubC). Patients with EC in the SubC group had the best survival and lowest ICD score, whereas patients in the SubB group had the worst prognosis. DEGs between subtypes were evaluated, and risk panels were established using LASSO-Cox regression analysis. The prognosis of low-risk patients was significantly better than that of high-risk patients in both cohorts. The area under the curve of the receiver operating characteristic curve indicated that the risk group had a good prognostic value. Our study identified the molecular subtypes of EC and ICD-based prognostic signatures. Our three-gene risk panel could serve as a biomarker for effectively assessing the prognostic risk of patients with EC.

5.
Neoplasia ; 38: 100888, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36804751

RESUMEN

Common sensitizing mutations in epidermal growth factor receptor (cEGFR), including exon 19 deletions (19-Del) and exon 21 L858R substitution, are associated with high sensitivity to EGFR-TKIs in NSCLC patients. The treatment for NSCLC patients with uncommon EGFR (uEGFR) mutations remains a subject of debate due to heterogeneity in treatment responses. In this manuscript, the targeted next-generation sequencing (NGS) data of a large cohort of EGFR-mutated NSCLC patients was assessed to elucidate genomic profiles of tumors carrying cEGFR or uEGFR mutations. The results showed that NSCLC patients with uEGFR mutations were more likely to harbor co-occurring genetic alterations in the Hippo pathway and a higher TMB compared with cEGFR-positive patients. Smoking-related mutations were found to significantly enriched in uEGFR-positive patients. Subgroup analyses were performed to identify potential prognostic biomarkers in patients harboring various EGFR subtype mutations. L858R-positive patients with co-existing ARID2 mutations had shorter progression-free survival (PFS) than those who were L858R- or 19-Del-positive but ARID2-negative (median: 2.3 vs. 12.0 vs. 8.0 months, P = 0.038). Furthermore, mutational profiles, such as top frequently mutated genes and mutational signatures of patients with various EGFR subtype mutations were significantly different. Our study analyzed the mutational landscape of NSCLC patients harboring cEGFR and uEGFR mutations, revealing specific genomic characteristics associated with uEGFR mutations that might explain the poor prognosis of first-generation EGFR-TKIs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Mutación , Receptores ErbB , Genómica
6.
J Exp Clin Cancer Res ; 42(1): 169, 2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37461053

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) contribute to multiple biological functions and are also involved in pathological conditions such as cancer. However, the role of circRNAs in metabolic reprogramming, especially upon energy stress in lung adenocarcinoma (LUAD), remains largely unknown. METHODS: Energy stress-induced circRNA was screened by circRNA profiling and glucose deprivation assays. RNA-seq, real-time cell analyzer system (RTCA) and measurement of oxygen consumption rate (OCR) were performed to explore the biological functions of circZFR in LUAD. The underlying mechanisms were investigated using circRNA pull-down, RNA immunoprecipitation, immunoprecipitation and bioinformatics analysis of alternative splicing. Clinical implications of circZFR were assessed in 92 pairs of LUAD tissues and adjacent non-tumor tissues, validated in established patient-derived tumor xenograft (PDTX) model. RESULTS: CircZFR is induced by glucose deprivation and is significantly upregulated in LUAD compared to adjacent non-tumor tissues, enhancing oxidative phosphorylation (OXPHOS) for adaptation to energy stress. CircZFR is strongly associated with higher T stage and poor prognosis in patients with LUAD. Mechanistically, circZFR protects heterogeneous nuclear ribonucleoprotein L-like (HNRNPLL) from degradation by ubiquitination to regulate alternative splicing, such as myosin IB (MYO1B), and subsequently activates the AKT-mTOR pathway to facilitate OXPHOS. CONCLUSION: Our study provides new insights into the role of circRNAs in anticancer metabolic therapies and expands our understanding of alternative splicing.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , MicroARNs , Humanos , ARN Circular/genética , ARN Circular/metabolismo , MicroARNs/genética , Fosforilación Oxidativa , Empalme Alternativo , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/patología , Regulación Neoplásica de la Expresión Génica , Proliferación Celular/genética , Línea Celular Tumoral
7.
Cancer Lett ; 577: 216426, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37820992

RESUMEN

The mechanisms underlying the involvement of long non-coding RNAs (lncRNAs) in the metastasis of small cell lung cancer (SCLC) remain largely unknown. Here, we identified that the lncRNA ITPR1-AS1 was upregulated in SCLC and lymph node metastasis tissues and positively correlated with SCLC malignant features. The overexpression of ITPR1-AS1 in SCLC was an independent risk factor for the overall survival of patients with SCLC. Our data confirmed that ITPR1-AS1 induces SCLC cell metastasis both in vitro and in vivo. Mechanistically, ITPR1-AS1 acts as a scaffold to enhance the interaction between SRC-associated in mitosis 68 kDa and heterogeneous nuclear ribonucleoprotein A1, which facilitates the alternative splicing of the H-Ras proto-oncogene (HRAS) pre-mRNA (P21HRAS). Moreover, we observed that ITPR1-AS1 could associate in a complex with and maintain the stability of DEAD-box polypeptide 3 (DDX3X), which inhibited the latter's ubiquitination and degradation. Our data provide evidence that ITPR1-AS1 activates the cRaf-MEK-ERK cascade by upregulating P21HRAS production and stabilizing DDX3X, to promote SCLC metastasis.


Asunto(s)
Neoplasias Pulmonares , ARN Largo no Codificante , Carcinoma Pulmonar de Células Pequeñas , Humanos , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , ARN Helicasas DEAD-box/genética , Regulación Neoplásica de la Expresión Génica , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Neoplasias Pulmonares/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Largo no Codificante/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo
8.
Adv Mater ; 34(15): e2110219, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35170096

RESUMEN

Tumorigenic environments, especially aberrantly overexpressed oncogenic microRNAs, play a critical role in various activities of tumor progression. However, developing strategies to effectively utilize and manipulate these oncogenic microRNAs for tumor therapy is still a challenge. To address this challenge, spherical nucleic acids (SNAs) consisting of gold nanoparticles in the core and antisense oligonucleotides as the shell are fabricated. Hybridized to the oligonucleotide shell is a DNA sequence to which doxorubicin is conjugated (DNA-DOX). The oligonucleotides shell is designed to capture overexpressed miR-21/miR-155 and inhibit the expression of these oncogenic miRNAs in tumor cells after tumor accumulation to manipulate genetic environment for accurate gene therapy. This process further induces the aggregation of these SNAs, which not only generates photothermal agents to achieve on-demand photothermal therapy in situ, but also enlarges the size of SNAs to enhance the retention time in the tumor for sustained therapy. The capture of the relevant miRNAs simultaneously triggers the intracellular release of the DNA-DOX from the SNAs to deliver tumor-specific chemotherapy. Both in vivo and in vitro results indicate that this combination strategy has excellent tumor inhibition properties with high survival rate of tumor-bearing mice, and can thus be a promising candidate for effective tumor treatment.


Asunto(s)
Nanopartículas del Metal , MicroARNs , Nanopartículas , Neoplasias , Ácidos Nucleicos , Animales , Carcinogénesis , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Oro , Nanopartículas del Metal/uso terapéutico , Ratones , MicroARNs/genética , Neoplasias/tratamiento farmacológico , Fototerapia
9.
ACS Appl Mater Interfaces ; 13(19): 23011-23019, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-33970619

RESUMEN

Remote controllability and multiple-shape memory performance are two important functions for shape memory polymers (SMPs) in engineering applications, which are still a challenge to achieve via a facile approach. Herein, we synthesized a shape memory composite with near-infrared (NIR) light-induced triple-shape memory performance by in situ formation of iron tannate (FeTA) nanoparticles in cross-linked poly(ethylene-co-vinyl alcohol) (EVOH). EVOH possessed two transition temperatures enabling the composites with triple-shape memory behavior, while FeTA nanoparticles served as the photothermal conversion factor for NIR light-induced responsiveness. Because the light-induced triple-shape memory performance of the composite is highly dependent on its photothermal conversion property, the control of FeTA doping would also be an effective solution to prepare light-induced multiple-SMPs with various shape transformations. Moreover, the composites exhibited high light-driving recovery stress, which could lift burdens 1600 times heavier than their own weight, indicating their great potential as a smart soft actuator for various applications.

10.
Acta Biomater ; 128: 408-419, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33878477

RESUMEN

Gene therapy based on transfection of RNAs/DNAs offers tremendous promise for tumor treatment. However, the relatively weak therapeutic efficiency of current genetic nanohybrids in vivo has limited the application of this strategy. Herein, we fabricated multifunctional core-shell-corona nanohybrids by combining cascaded theranostics for enhanced gene therapy. The nanohybrids consist of polydopamine-modified Fe3O4 nanoparticles as core, anti-miRNA-21 oligonucleotides (anti-miRNA) strands as shell, and doxorubicin (DOX)-conjugated DNA-8pb (DOX-DNA-8bp) as corona. The polydopamine/Fe3O4 core not only serves as an active agent for local photothermal therapy under NIR irradiation, but it also provides magnetic targeting to tumor tissue for accurate treatment, which could enhance the therapeutic effect and reduce the undesired side effects to healthy tissues. The DOX-DNA-8bp corona was grafted on the anti-miRNA shell through base pairing, which could be replaced by overexpressed miRNA-21 in tumor cells due to the strong interaction between miRNA-21 and anti-miRNA, resulting in tumor-specific gene therapy through tumorigenic miRNA-21 consumption and tumor selective chemotherapy through miRNA-21-triggered DOX-DNA-8bp release in tumor cells. Moreover, the intelligent controlled release system can gradually stop the release of DOX to prevent side effects caused by drug overdose, once sufficient damage of tumor cells has occurred, due to the downregulation of miRNA-21. The results of both in vitro and in vivo analyses showed that the nanohybrids combining cascaded chemo-photo-gene therapy could effectively inhibit tumor growth, promote the survival of tumor-bearing mice, and show no visible adverse effects on these mice, resulting in a promising nanoplatform for tumor treatment. STATEMENT OF SIGNIFICANCE: Gene therapy based on transfection of RNAs/DNAs offers tremendous promise for cancer treatment. However, the relatively weak therapeutic efficiency of current genetic nanovectors in vivo that results in insufficient tumor targeting and easy decomposition/elimination of RNAs/DNAs during therapy has limited its application. Although some approaches have combined photothermal agents or antitumor drugs with RNA/DNA nanocarriers to achieve better treatment, the spatiotemporal differences in photothermal therapy, chemotherapy, and gene therapy using current nanohybrids may hinder their synergistic effect. In the present study, we fabricated multifunctional core-shell-corona nanohybrids (Fe3O4@PDA@anti-miRNA/DNA) to simultaneously perform on-demand photothermal therapy, miR-21-triggered chemotherapy, and miR-21-dependent gene therapy at the same location, which can achieve an efficient synergistic effect for precise and effective tumor treatment.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Animales , Doxorrubicina/farmacología , Ratones , Fototerapia , Medicina de Precisión
11.
Biomater Sci ; 9(15): 5237-5246, 2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34223579

RESUMEN

The combination of therapeutic and diagnostic functions in a single platform has aroused great interest due to the more optimal synergistic effects that can be obtained as compared to any single theranostic approach alone. However, current nanotheranostics are normally formed via complicated construction steps involving the pre-synthesis of each component and further conjugation via chemical bonds, which may cause low integration efficiency and limit production and applications. Herein, a tumor-targeting and tumor-responsive all-in-one nanoplatform based on mesoporous silica nanocarriers (MSNs) was fabricated via a facile approach utilizing efficient and nondestructive physical interactions for long-wavelength fluorescence imaging-guided synergistic chemo-catalytic-photothermal tumor therapy. The MSNs were endowed with these multimodal theranostics via a simple hydrothermal method after coordinating with Fe2+ and glutathione (GSH) to introduce ferroferric oxide and carbon dots in situ. The former acts as a photothermal agent and catalytic agent to generate local heat under 808 nm irradiation and also when toxic hydroxyl radicals (˙OH) are in contact with abundant hydrogen peroxide in cancer cells, while the latter participates in fluorescence imaging. After loading with paclitaxel (PTX), polyester and folic-acid-conjugated cyclodextrin were employed to serve as an esterase-sensitive gatekeeper controlling PTX release from the MSN pores and as a tumor-targeting agent for accurate therapy, respectively. As expected, the nanoplatform was efficiently taken up by tumor cells over healthy cells, and then, synergetic chemo-catalytic-photothermal therapy was performed, resulting in 5-fold greater apoptosis of tumor cells as compared to healthy cells under 808 nm irradiation. Moreover, in vivo data from tumor-bearing mouse models showed that tumors were significantly inhibited, and the survival rates of these mice increased to greater than 80% after 5 weeks of treatment with our nanoplatform. These therapeutic processes could be directly tracked via fluorescence imaging enabled by carbon dots and, therefore, our nanoplatform provides a promising theranostics approach for tumor treatment.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Doxorrubicina , Ratones , Paclitaxel , Medicina de Precisión , Dióxido de Silicio
12.
Biomaterials ; 278: 121181, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34653932

RESUMEN

Sufficient energy generation based on effective transport of nutrient via abundant blood vessels in tumor tissue and subsequent oxidative metabolism in mitochondria is critical for growth, proliferation and migration of tumor. Thus the strategy to cut off this transport pathway (blood vessels) and simultaneously close the power house (mitochondria) is highly desired for tumor treatment. Herein, we fabricated a bionic nanocarrier with core-shell-corona structure to give selective and effective tumor therapy via stepwise destruction of existed tumor vessel, inhibition of tumor angiogenesis and dysfunction of tumor mitochondria. The core of this bionic nanocarrier consists of combretastatin A4 phosphate (CA4P) and vitamin K2 (VK2) co-loaded mesoporous silica nanoparticle (MSNs), which is in charge of the vasculature destruction and mitochondrial dysfunction after cargos release. The N-tert-butylacrylamide (TBAM) and tri-sulfated N-acetylglucosamine (TSAG) shell served as artificial affinity reagent against vascular endothelial growth factor (VEGF) for angiogenesis inhibition. As to guarantee that these actions only happened in tumor, the hyaluronic acid (HA) corona was introduced to endow the nanocarrier with tumor targeting property and stimuli-responsiveness for accurate therapy. Both in vitro and in vivo results indicated that the CA4P/VK2-MSNs-TBAM/TSAG-HA (CVMMGH for short) nanocarrier combined well-controllable manipulation of tumor vasculature and tumor mitochondria to effectivly cut off the tumorigenic energy supply, which performed significant inhibition of tumor growth, demonstrating the great candidate of our strategy for effective tumor therapy.


Asunto(s)
Nanopartículas , Neoplasias , Biónica , Humanos , Neoplasias/tratamiento farmacológico , Dióxido de Silicio , Factor A de Crecimiento Endotelial Vascular
13.
Biomaterials ; 275: 120956, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34146890

RESUMEN

Proper manipulation of tumorigenic microenvironments has been considered as one of the most effective approaches for tumor therapy, which is still a challenge to be well performed. Herein, a nano-modulator was fabricated to manipulate the hypoxia, glucose, radicals and local temperature in tumor tissue as needed, which consists of hemoglobin (Hb) and ferric ion (Fe3+) co-conjugated polydopamine (PDA) as core, glucose oxidase (GOD) as shell, and folic acid (FA) modified polyethylene glycol (PEG) as corona. The PEG-FA corona not only protected Hb and GOD against protease in blood circulation, but serve as tumor targeting agent for tumor specific accumulation of the nano-modulator. The Hb is in charge of oxygen supply to reverse the hypoxic environment of tumor tissue, which promotes the function of GOD to achieve rapid glucose consumption and hydrogen peroxide generation. The polydopamine was employed to raise local temperature under NIR irradiation, meanwhile to continuously reduce Fe3+ to produce ferrous ions (Fe2+), which further catalyze hydrogen peroxide to cytotoxic hydroxyl radicals via Fenton reaction. Both in vitro and in vivo results showed excellent tumor inhibition and high survival rate of tumor-bearing mice after treatment by our nano-modulator, indicating this synergistic therapy via on-demand manipulation of various tumorigenic microenvironments could be a green approach for tumor treatment with high efficiency and minimum side effects.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Catálisis , Línea Celular Tumoral , Glucosa Oxidasa , Peróxido de Hidrógeno , Ratones , Neoplasias/tratamiento farmacológico , Polietilenglicoles , Microambiente Tumoral
14.
Sci Transl Med ; 13(584)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692135

RESUMEN

The therapeutic goal for autoimmune diseases is disease antigen-specific immune tolerance without nonspecific immune suppression. However, it is a challenge to induce antigen-specific immune tolerance in a dysregulated immune system. In this study, we developed immune-homeostatic microparticles (IHMs) that treat multiple mouse models of autoimmunity via induction of apoptosis in activated T cells and reestablishment of regulatory T cells. Specifically, in an experimental model of colitis, IHMs rapidly released monocyte chemotactic protein-1 after intravenous administration, which recruited activated T cells and then induced their apoptosis by conjugated Fas ligand on the IHM surface. This triggered professional macrophages to ingest apoptotic T cells and produce high quantities of transforming growth factor-ß, which drove regulatory T cell differentiation. Furthermore, the modular design of IHMs allowed IHMs to be engineered with the autoantigen peptides that can reduce disease in an experimental autoimmune encephalomyelitis mouse model and a nonobese diabetic mouse model. This was accomplished by sustained release of the autoantigens after induction of T cell apoptosis and transforming growth factor-ß production by macrophages, which promoted to establish an immune tolerant environment. Thus, IHMs may be an efficient therapeutic strategy for autoimmune diseases through induction of apoptosis and reestablishment of tolerant immune responses.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Tolerancia Inmunológica , Animales , Autoantígenos , Autoinmunidad , Ratones , Linfocitos T Reguladores
15.
Cancer Commun (Lond) ; 41(6): 492-510, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33734616

RESUMEN

BACKGROUND: Considering the increase in the proportion of lung adenocarcinoma (LUAD) cases among all lung cancers and its considerable contribution to cancer-related deaths worldwide, we sought to identify novel oncogenes to provide potential targets and facilitate a better understanding of the malignant progression of LUAD. METHODS: The results from the screening of transcriptome and survival analyses according to the integrated Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) data were combined, and a promising risk biomarker called meiotic nuclear divisions 1 (MND1) was selectively acquired. Cell viability assays and subcutaneous xenograft models were used to validate the oncogenic role of MND1 in LUAD cell proliferation and tumor growth. A series of assays, including mass spectrometry, co-immunoprecipitation (Co-IP), and chromatin immunoprecipitation (ChIP), were performed to explore the underlying mechanism. RESULTS: MND1 up-regulation was identified to be an independent risk factor for overall survival in LUAD patients evaluated by both tissue microarray staining and third party data analysis. In vivo and in vitro assays showed that MND1 promoted LUAD cell proliferation by regulating cell cycle. The results of the Co-IP, ChIP and dual-luciferase reporter assays validated that MND1 competitively bound to tumor suppressor Kruppel-like factor 6 (KLF6), and thereby protecting E2F transcription factor 1 (E2F1) from KLF6-induced transcriptional repression. Luciferase reporter and ChIP assays found that E2F1 activated MND1 transcription by binding to its promoter in a feedback manner. CONCLUSIONS: MND1, KLF6, and E2F1 form a positive feedback loop to regulate cell cycle and confer DDP resistance in LUAD. MND1 is crucial for malignant progression and may be a potential therapeutic target in LUAD patients.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular , División del Núcleo Celular , Factor de Transcripción E2F1/genética , Retroalimentación , Humanos , Factor 6 Similar a Kruppel , Neoplasias Pulmonares/genética
16.
Genes (Basel) ; 11(11)2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33158173

RESUMEN

Metastasis and recurrence are the main causes of lung adenocarcinoma patients' death. Lymphatic metastasis is the main way of non-small cell lung cancer (NSCLC) metastasis. C-C chemokine receptor type 7 (CCR7) overexpression has been demonstrated to mediate occurrence and progression of NSCLC. Moreover, Chemokine ligand 21 (CCL21) was used to activate CCR7. The CCR7-CCL21 axis is one of the most common "chemokine-receptor" modes of action in the development and metastasis of multiple tumors. However, the role of the CCR7-CCL21 axis in lymphatic metastasis of NSCLC is poorly understood. The study was conducted to investigate the molecular mechanism underlying CCR7-CCL21 axis-mediated lymphatic metastasis of NSCLC A549 cells. Tumor necrosis factor α (TNF-α) could regulate the tumor microenvironment balance by promoting chemokine secretion. Our study demonstrated that TNF-α promoted CCL21 production in human lymphatic endothelial cells (HLEC). Results further showed that TNF-α significantly activated the NF-κB pathway in HLEC. NF-κB pathway inhibition with ammonium pyrrolidinedithiocarbamate (PDTC) caused a significant decrease in CCL21 secretion, suggesting that TNF-α-induced CCL21 secretion in HLEC was through NF-κB pathway. Co-culture of A549 cells and TNF-α-treated HLEC confirmed that the metastasis of A549 cells was enhanced, meanwhile, apoptosis-related proteins were hardly affected. The data proved that a co-culture system prevented cell apoptosis while inducing the lymphatic metastasis of A549 cells. However, the situation was reversed after neutralizing CCL21 expression, suggesting that TNF-α-induced CCL21 secretion in HLEC is involved in A549 cells metastasis. Collectively, our finding demonstrated that NF-κB pathway-controlled CCL21 secretion of HLEC contributing to the lymphatic metastasis of A549 cells via the CCR7-CCL21 axis, validating the CCR7-CCL21 axis as a potential target to inhibit metastasis of NSCLC.


Asunto(s)
Quimiocina CCL21/genética , Metástasis Linfática/genética , Receptores CCR7/genética , Células A549 , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CCL21/metabolismo , Quimiotaxis/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Neoplasias Pulmonares/metabolismo , Metástasis Linfática/fisiopatología , FN-kappa B/metabolismo , Recurrencia Local de Neoplasia/genética , Receptores CCR7/metabolismo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/fisiología
17.
Biomaterials ; 262: 120357, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32911253

RESUMEN

Chemotherapy is one of the most commonly utilized approaches to treat malignant tumor. However, the well-controlled chemotherapy able to accurately manipulate local drugs for on-demand tumor treatment is still a challenge. Herein, a magnet and light dual-responsive hydrogel combining thermosensitive poly(N-acryloyl glycinamide) (PNAGA), doxorubicin (DOX) loaded and polyester (PE) capped mesoporous silica nanocarriers (MSNs) as well as Fe3O4 nanoparticles (Fe3O4 NPs) grafted graphene oxide (GO) was fabricated to address above issue. The Fe3O4 NPs and GO respectively serve as magnetothermal agent and photothermal agent to perform hyperthermia, meanwhile to generate chain motion of PNAGA with varying degrees under different conditions of magnetic field and/or NIR irradiation. This strategy not only allowed the gel-sol transition of the hydrogel by prior heating for tumor injection, but performed controllable release routes of DOX-MSNs-PE (DMP for short) nanocarriers to meet various requirements from different patients and the changing states of tumor. Furthermore, these escaped DMP nanocarriers could be taken by surrounding tumor cells, and then deliver their drug to these cells after rapid hydrolysis of the PE cap triggered by esterase, resulting in accurate chemotherapy. Both in vitro and in vivo results indicated that the PNAGA-DMP-Fe3O4@GO hydrogel combining well-controllable chemotherapy and hyperthermia can eliminate more than 90% tumor cells and effectively inhibit the tumor growth in mice model, demonstrating the great candidate of our hydrogel for accurate tumor therapy.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Animales , Doxorrubicina , Humanos , Imanes , Ratones , Nanogeles , Dióxido de Silicio
18.
J Mater Chem B ; 8(31): 6857-6865, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32022092

RESUMEN

Catalytic medicine based on various catalysts has attracted increasing interest for the treatment of tumors. However, the direct application of conventional catalysts may cause serious side effects to healthy tissue and low therapeutic efficiency against tumor tissue, due to their weak specificity for the tumor microenvironment (TME). Herein, a tumor-targeting and TME-responsive nanoreactor containing ferroferric oxide nanoparticles (Fe3O4 NPs) and glucose oxidase (GOD) was developed to perform hyaluronidase (HAase) and glutathione (GSH)-triggered chain catalytic reactions in tumor tissue. This nanoreactor was designed to take advantage of the unique biological molecules in tumors and several therapeutic agents to adjust the local microenvironments, and achieved satisfactory and accurate tumor therapy. The reactions started with the consumption of intratumoral glucose to inhibit tumor growth, and simultaneously produced hydrogen peroxide (H2O2) to make up for the deficiency of H2O2 in the original tumor microenvironment, resulting in the generation of a high quantity of hydroxyl radicals as a result of the catalysis of Fe3O4 NPs to further eliminate tumor tissue. The tumor-specific catalytic medicine formed by our nanocomposite guaranteed both therapeutic efficiency and accuracy, avoiding potential risks to healthy tissue and leading to a four-fold improvement in the cytotoxicity against tumor cells compared with normal cells after incubations of 48 h. In vivo data from mouse models provided further evidence for its therapeutic efficacy: the tumor growth was completely inhibited after two weeks of the synergistic therapy, which indicated the promise of our nanocomposite for tumor treatment.


Asunto(s)
Biocatálisis , Nanomedicina/métodos , Animales , Línea Celular Tumoral , Proliferación Celular , Glucosa Oxidasa/metabolismo , Glutatión/metabolismo , Humanos , Hialuronoglucosaminidasa/metabolismo , Radical Hidroxilo/metabolismo , Nanopartículas Magnéticas de Óxido de Hierro/química , Ratones , Microambiente Tumoral
19.
Thorac Cancer ; 11(8): 2137-2145, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32567804

RESUMEN

BACKGROUND: Lung adenocarcinoma (LUAD) is the most predominant histological type of lung cancer with a poor prognosis. In this study, we demonstrate that LBX2 regulates cell proliferation, migration and invasion and the potential molecular mechanism in LUAD. METHODS: The Cancer Genome Atlas dataset was accessed to screen for novel genes and immunohistochemistry (IHC) assays were performed to determine the association between LBX2 expression and clinicopathological features of LUAD. 5-ethynyl-2'-deoxyuridine, colony formation and Real Time xCELLigence analysis system were used to evaluate the cell proliferation abilities of LUAD. Wound healing, transwell and Matrigel assays were used to detect cell migration and invasion capacities. Xenograft tumor models were used to assess the oncogenic role of LBX2 in vivo. RESULTS: We found that LBX2 was hyperexpressed in LUAD and correlated with clinicopathological features and poor prognosis in LUAD patients. Knockdown of LBX2 inhibited cell proliferation, migration and invasion of LUAD, whereas ectopic expression of LBX2 enhanced tumor growth, migration, and invasion. We further found that LBX2 might participate in epithelial-to-mesenchymal transition (EMT) progression and influence EMT-related gene expression. CONCLUSIONS: The current study suggests that LBX2 plays an oncogenic role in LUAD and may participate in tumor proliferation, migration, and invasion through EMT progression. KEY POINTS: Significant findings of the study LBX2 might participate in LUAD cell proliferation, migration and invasion via EMT progression. What this study adds LBX2 may represent a potential biomarker and a promising therapeutic target for LUAD.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Animales , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Transfección
20.
J Mater Chem B ; 8(34): 7777-7784, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32744264

RESUMEN

Tissue fibrosis caused by implantation of tissue engineering scaffolds is an urgent problem in clinical research. In this work, a substrate-independent coating with on-demand release of an antifibrotic drug has been fabricated to effectively address this issue. This coating was formed through a substrate-independent layer-by-layer (LBL) technique via a cationic polyelectrolyte (poly-diallyldimethylammonium, PDDA) and an anionic polyelectrolyte (poly-styrenesulfonate, PSS), where parts of PSS and PDDA were physically replaced by carboxyl functionalized polyethylene glycol grafted onto antifibrotic drug dexamethasone (DEX-PEG-COOH). Considering the easy generation of local inflammation after implantation, an ester bond was designed between PEG-COOH and DEX. Therefore, the overexpressed esterase under inflammatory conditions hydrolyzes the ester bond and thereby releases DEX from the film to inhibit fibrosis occurring in the tissue repair process. The in vivo capacity of this coating to restrain tissue fibrosis was investigated by a skin defect model using porous polycaprolactone (PCL) scaffolds as substrates. The experimental results showed that the fibrosis-related proteins (Col-I, TGF-ß and fibronectin) and the infiltration of myofibroblasts (α-SMA) of skin tissues in the coated PCL scaffold group were significantly lower than those in the blank control group and pure PCL scaffold group. Moreover, the histological evaluations showed that the coating group could significantly decrease the deposition of collagen and meanwhile promote the partial regeneration of skin appendages. These results successfully demonstrate that the universal coating prepared with a simple protocol would be an effective strategy to address the fibrosis issues during tissue engineering.


Asunto(s)
Dexametasona/química , Dexametasona/farmacología , Portadores de Fármacos/química , Liberación de Fármacos , Polímeros/química , Colágeno/metabolismo , Esterificación , Fibrosis , Humanos , Hidrólisis , Poliésteres/química , Porosidad , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA