Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
ACS Nano ; 18(40): 27557-27569, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39329362

RESUMEN

Radiation therapy, a fundamental treatment for tumors, is often accompanied by radiation-induced skin injury (RISI). Excessive production of reactive oxygen species (ROS) and subsequent inflammation are two key factors in RISI development that will cause skin injury and affect radiotherapy. Herein, the co-assembled binary polyphenol natural products inspired the development of a dual-functional cascade microneedle system for prevention and treatment of RISI. Specifically, epigallocatechin gallate (EGCG) and curcumin (CUR) were co-assembled into nanoparticles (CEPG) by intermolecular interactions and then incorporated with catalase (CAT) to achieve a cascade system in the microneedles (this microneedle system was conducive to penetrate into the epidermal keratinocytes where RISI had the greatest impact). When using microneedles, the tip dissolved rapidly and delivered CEPG and CAT into the dermis, where CEPG NPs were able to respond to ROS and decompose into EGCG and CUR. More importantly, EGCG and CAT formed a cascade that converts superoxide anions into water step-by-step, which can reduce cell damage caused by free radicals in the early stages of radiation for prevention; meanwhile, CUR inhibited inflammatory pathways, achieving the treatment of skin inflammation in the post-radiotherapy period. These explorations broaden the strategy for the application of natural products in RISI.


Asunto(s)
Productos Biológicos , Catequina , Polifenoles , Productos Biológicos/farmacología , Productos Biológicos/química , Animales , Polifenoles/farmacología , Polifenoles/química , Catequina/farmacología , Catequina/análogos & derivados , Catequina/química , Humanos , Piel/efectos de los fármacos , Piel/patología , Piel/efectos de la radiación , Curcumina/farmacología , Curcumina/química , Ratones , Especies Reactivas de Oxígeno/metabolismo , Nanopartículas/química , Catalasa/metabolismo , Traumatismos por Radiación/prevención & control , Traumatismos por Radiación/tratamiento farmacológico
3.
J Mater Chem B ; 12(26): 6466-6479, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38864401

RESUMEN

The urgent need to curb the rampant rise in cancer has impelled the rapid development of nanomedicine. Under the above issue, transition metal compounds have received special attention considering their physicochemical and biochemical properties. However, how to take full advantage of the valuable characteristics of nanomaterials based on their spatial structures and chemical components for synergistic tumor therapy is a worthwhile exploration. In this work, a tailored two-dimensional (2D) FeSe2 nanosheet (NS) platform is proposed, which integrates enzyme activity and drug efficacy through the regulation of itsstability. Specifically, metastable FeSe2 NSs can serve as dual nanozymes in an intact state, depleting GSH and increasing ROS to induce oxidative stress in the tumor microenvironment (TME). With the gradual degradation of the FeSe2 in TME, its degraded products can amplify the Fenton reaction and GSH consumption, enhance the expression of inflammatory factors, and achieve effective near-infrared (NIR)-light irradiation-enhanced synergistic photothermal therapy (PTT) and chemodynamic therapy (CDT). Our exploration further confirmed such a strategy that may integrate carrier activity and drug action into a metastable nanoplatform for tumor synergistic therapy. These results prompt the consideration of the rational design of a one-for-all carrier that can exhibit multifunctional properties and nanomedicine efficacy for versatile therapeutic applications in the future.


Asunto(s)
Antineoplásicos , Nanoestructuras , Animales , Ratones , Nanoestructuras/química , Antineoplásicos/química , Antineoplásicos/farmacología , Humanos , Microambiente Tumoral/efectos de los fármacos , Terapia Fototérmica , Proliferación Celular/efectos de los fármacos , Tamaño de la Partícula , Ensayos de Selección de Medicamentos Antitumorales , Ratones Endogámicos BALB C , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Neoplasias/tratamiento farmacológico
4.
Free Radic Biol Med ; 222: 244-258, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38901499

RESUMEN

Doxorubicin (Dox) use is limited by Dox-induced cardiotoxicity. TANK-blinding kinase 1 (TBK1) is an important kinase involved in the regulation of mitophagy, but the role of TBK1 in cardiomyocytes in chronic Dox-induced cardiomyopathy remains unclear. Cardiomyocyte-specific Tbk1 knockout (Tbk1CKO) mice received Dox (6 mg/kg, injected intraperitoneally) once a week for 4 times, and cardiac assessment was performed 4 weeks after the final Dox injection. Adenoviruses encoding Tbk1 or containing shRNA targeting Tbk1, or a TBK1 phosphorylation inhibitor were used for overexpression or knockdown of Tbk1, or inhibit phosphorylation of TBK1 in isolated primary cardiomyocytes. Our results revealed that moderate Dox challenge decreased TBK1 phosphorylation (with no effect on TBK1 protein levels), resulting in compromised myocardial function, obvious mortality and overt interstitial fibrosis, and the effects were accentuated by Tbk1 deletion. Dox provoked mitochondrial membrane potential collapse and oxidative stress, the effects of which were exacerbated and mitigated by Tbk1 knockdown, specific inhibition of phosphorylation and overexpression, respectively. However, Tbk1 (Ser172A) overexpression did not alleviate these effects. Further scrutiny revealed that TBK1 exerted protective effects on mitochondria via SQSTM1/P62-mediated mitophagy. Tbk1 overexpression mediated cardioprotective effects on Dox-induced cardiotoxicity were cancelled off by Sqstm1/P62 knockdown. Moreover, TBK1-mitophagy-mitochondria cascade was confirmed in heart tissues from dilated cardiomyopathy patients. Taken together, our findings denoted a pivotal role of TBK1 in Dox-induced mitochondrial injury and cardiotoxicity possibly through its phosphorylation and SQSTM1/P62-mediated mitophagy.


Asunto(s)
Cardiotoxicidad , Doxorrubicina , Ratones Noqueados , Mitofagia , Miocitos Cardíacos , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/efectos de los fármacos , Mitofagia/efectos de los fármacos , Mitofagia/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Doxorrubicina/efectos adversos , Ratones , Cardiotoxicidad/genética , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , Cardiotoxicidad/etiología , Estrés Oxidativo/efectos de los fármacos , Humanos , Fosforilación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Masculino
5.
J Transl Med ; 22(1): 297, 2024 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-38515161

RESUMEN

BACKGROUND: The aberrant secretion and excessive deposition of type I collagen (Col1) are important factors in the pathogenesis of myocardial fibrosis in dilated cardiomyopathy (DCM). However, the precise molecular mechanisms underlying the synthesis and secretion of Col1 remain unclear. METHODS AND RESULTS: RNA-sequencing analysis revealed an increased HtrA serine peptidase 1 (HTRA1) expression in patients with DCM, which is strongly correlated with myocardial fibrosis. Consistent findings were observed in both human and mouse tissues by immunoblotting, quantitative reverse transcription polymerase chain reaction (qRT-PCR), immunohistochemistry, and immunofluorescence analyses. Pearson's analysis showed a markedly positive correlation between HTRA1 level and myocardial fibrosis indicators, including extracellular volume fraction (ECV), native T1, and late gadolinium enhancement (LGE), in patients with DCM. In vitro experiments showed that the suppression of HTRA1 inhibited the conversion of cardiac fibroblasts into myofibroblasts and decreased Col1 secretion. Further investigations identified the role of HTRA1 in promoting the formation of endoplasmic reticulum (ER) exit sites, which facilitated the transportation of Col1 from the ER to the Golgi apparatus, thereby increasing its secretion. Conversely, HTRA1 knockdown impeded the retention of Col1 in the ER, triggering ER stress and subsequent induction of ER autophagy to degrade misfolded Col1 and maintain ER homeostasis. In vivo experiments using adeno-associated virus-serotype 9-shHTRA1-green fluorescent protein (AAV9-shHTRA1-GFP) showed that HTRA1 knockdown effectively suppressed myocardial fibrosis and improved left ventricular function in mice with DCM. CONCLUSIONS: The findings of this study provide valuable insights regarding the treatment of DCM-associated myocardial fibrosis and highlight the therapeutic potential of targeting HTRA1-mediated collagen secretion.


Asunto(s)
Cardiomiopatías , Cardiomiopatía Dilatada , Animales , Humanos , Ratones , Colágeno Tipo I , Medios de Contraste , Fibrosis , Gadolinio , Miocardio/patología
6.
Front Immunol ; 15: 1341209, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38352864

RESUMEN

Background: Aminooctylamine (ANO1) plays an oncogenic role in various cancers. However. its role in pancreatic cancer (PC) has rarely been studied. This study investigated the prognostic value of ANO1 and its correlation with the tumor microenvironment (TME) in PC. Methods: Consecutive patients with PC (n = 119) were enrolled. The expression of ANO1 in cancer cells, the expression of fibroblast activation protein (FAP) and alpha smooth muscle actin in cancer-associated fibroblasts (CAFs), and the numbers of CD8- and FOXP3-positive tumor-infiltrating lymphocytes (TILs) were evaluated using immunohistochemistry. The prognostic value of ANO1 and its correlation with CAF subgroups and TILs were analyzed. The possible mechanism of ANO1 in the TME of PC was predicted using the the Cancer Genome Atlas (TCGA) dataset. Results: The expression of AN01 was correlated with overall survival (OS) and disease-free survival. Multi-factor analysis showed that high ANO1 expression was an independent adverse prognostic factor for OS (hazard ratio, 4.137; P = 0.001). ANO1 expression was positively correlated with the expression of FAP in CAFs (P < 0.001) and negatively correlated with the number of CD8-positive TILs (P = 0.005), which was also validated by bioinformatics analysis in the TCGA dataset. Moreover, bioinformatic analysis of the TCGA dataset revealed that ANO1 may induce an immunosuppressive tumor microenvironment in pancreatic cancer in a paracrine manner. Conclusion: ANO1 is a prognostic factor in patients with PC after radical resection. ANO1 may induce an immunosuppressive tumor microenvironment in PC in a paracrine manner, suggesting that ANO1 may be a novel therapeutic target.


Asunto(s)
Neoplasias Pancreáticas , Microambiente Tumoral , Humanos , Pronóstico , Neoplasias Pancreáticas/patología , Linfocitos Infiltrantes de Tumor/metabolismo , Modelos de Riesgos Proporcionales , Anoctamina-1/genética , Anoctamina-1/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
7.
Adv Healthc Mater ; 13(6): e2302721, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37990787

RESUMEN

Multimodal biomedical imaging and imaging-guided therapy have garnered extensive attention owing to the aid of nanoagents with the aim of further improving the therapeutic efficacy of diseases. The ability to engineer nanocomplexes (NCs) or control how they behave within an organism remains largely elusive. Here, a multifunctional nanoplatform is developed based on stabilized I-doped perovskite, CsPbBr3 -x Ix @SiO2 @Lip-c(RGD)2 (PSL-c(RGD)2 ) NCs. In particular, by regulating the amount of regular I- ions introduced, the fluorescence emission spectrum of perovskite-based NCs can be modulated well to match the requirement for biomedical optical imaging at the scale from molecule, cell to mouse; doping 125 I enables the nanoformulation to be competent for single-photon emission computed tomography (SPECT) imaging; the introduction of 131 I- imparts the NCs with the capability for radiotherapy. Through facile manipulation of specific iodine ions, this nanoplatform exhibits a remarkable ability to match multifunctional biomedical imaging and tumor therapy. In addition, their in vivo behavior can be manipulated by adjusting the thickness of the silica shell and the surface polarity for more practical applications. These experimental explorations offer a novel approach for engineering desirable multimodal NCs to simultaneously image and combat malignant tumors.


Asunto(s)
Compuestos de Calcio , Radioisótopos de Yodo , Yodo , Neoplasias , Óxidos , Titanio , Animales , Ratones , Dióxido de Silicio , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia , Iones , Oligopéptidos
8.
ACS Appl Mater Interfaces ; 15(23): 27612-27623, 2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37265327

RESUMEN

The extensive research into developing novel strategies for detecting respiratory syndrome coronavirus 2 (SARS-CoV-2) antigens in clinical specimens, especially the sensitive point-of-care testing method, is still urgently needed to reach rapid screening of viral infections. Herein, a new lateral flow immunoassay (LFIA) platform was reported for the detection of SARS-CoV-2 spike-S1 protein antigens, in which four sensitive and specific SARS-CoV-2 mouse monoclonal antibodies (MmAbs) were tailored by using quantum dot (QD)-loaded dendritic mesoporous silica nanoparticles modified further for achieving the -COOH group surface coating (named Q/S-COOH nanospheres). Importantly, compact QD adsorption was achieved in mesoporous channels of silica nanoparticles on account of highly accessible central-radial pores and electrostatic interactions, leading to significant signal amplification. As such, a limit of detection for SARS-CoV-2 spike-S1 testing was found to be 0.03 ng/mL, which is lower compared with those of AuNPs-LFIA (traditional colloidal gold nanoparticles, Au NPs) and enzyme-linked immunosorbent assay methods. These results show that optimizing the affinity of antibody and the intensity of fluorescent nanospheres simultaneously is of great significance to improve the sensitivity of LFIA.


Asunto(s)
COVID-19 , Nanopartículas del Metal , Nanosferas , Animales , Ratones , SARS-CoV-2 , COVID-19/diagnóstico , Oro , Dióxido de Silicio , Inmunoensayo/métodos , Anticuerpos Antivirales , Sensibilidad y Especificidad
9.
J Mater Chem B ; 11(20): 4498-4510, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37159229

RESUMEN

Various unique spatial structures are often found in the enzymes of biological systems. From the consideration of bionics, it is challenging but meaningful to design nanozymes with distinctive structures to enhance their bioactivities. To explore the relationship between the structure and activity of nanozymes, in this work, a special structural nanoreactor, namely small pore black TiO2 coated/doped large pore Fe3O4 (TiO2/-Fe3O4) loaded with lactate oxidase (LOD), was constructed for chemodynamic and photothermal synergistic therapy. Specifically, LOD loaded on the surface of the TiO2/-Fe3O4 nanozyme alleviates the low level of H2O2 in the tumour microenvironment (TME); the black TiO2 shell with multiple pinhole channels and a large specific surface area not only facilitates LOD loading, but also enhances the affinity of the nanozyme for H2O2; H2O2 is continuously enriched on the surface of the TiO2/-Fe3O4 nanozyme and transmitted to mesoporous Fe3O4, in turn efficiently producing abundant toxic hydroxyl radicals (˙OH) for chemodynamic therapy. Meanwhile, the TiO2/-Fe3O4 nanozyme under 1120 nm laser irradiation has excellent photothermal conversion efficiency (η = 41.9%), and further accelerates the production of ˙OH for amplifying the chemodynamic therapy efficiency. This self-cascading, special structure nanozyme provides a novel strategy for application in highly efficient tumour synergetic therapy.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Humanos , Peróxido de Hidrógeno/farmacología , Biónica , Nanotecnología , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
10.
Sci Rep ; 13(1): 6622, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37095256

RESUMEN

The tumour microenvironment (TME) is vital to tumour development and influences the immunotherapy response. Abnormal nucleotide metabolism (NM) not only promotes tumour cell proliferation but also inhibits immune responses in the TME. Therefore, this study aimed to determine whether the combined signatures of NM and the TME could better predict the prognosis and treatment response in gastric cancer (GC). 97 NM-related genes and 22 TME cells were evaluated in TCGA-STAD samples, and predictive NM and TME characteristics were determined. Subsequent correlation analysis and single-cell data analysis illustrated a link between NM scores and TME cells. Thereafter, NM and TME characteristics were combined to construct an NM-TME classifier. Patients in the NMlow/TMEhigh group exhibited better clinical outcomes and treatment responses, which could be attributed to the differences in immune cell infiltration, immune checkpoint genes, tumour somatic mutations, immunophenoscore, immunotherapy response rate and proteomap. Additionally, the NMhigh/TMElow group benefited more from Imatinib, Midostaurin and Linsitinib, while patients in the NMlow/TMEhigh group benefited more from Paclitaxel, Methotrexate and Camptothecin. Finally, a highly reliable nomogram was developed. In conclusion, the NM-TME classifier demonstrated a pretreatment predictive value for prognosis and therapeutic responses, which may offer novel strategies for strategizing patients with optimal therapies.


Asunto(s)
Neoplasias Gástricas , Humanos , Pronóstico , Microambiente Tumoral , Biomarcadores , Inmunoterapia
11.
Int Immunopharmacol ; 118: 110070, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37003186

RESUMEN

Rationale Idiopathic pulmonary fibrosis (IPF) is a lung disease with high mortality, limited treatment options and an unknown aetiology. M2 macrophages play a critical role in the pathological process of IPF. Triggering receptor expressed on myeloid cells-2 (TREM2) participates in the regulation of macrophages, although its role in IPF remains elusive. METHODS: This study examined the role of TREM2 in macrophage regulation using a well-established bleomycin (BLM)-induced pulmonary fibrosis (PF) mouse model. TREM2 insufficiency was induced by intratracheal treatment with TREM2-specific siRNA. The effects of TREM2 on IPF were evaluated using histological staining and molecular biological methods. RESULTS: TREM2 expression levels were significantly elevated in the lungs of IPF patients and mice with BLM-induced pulmonary fibrosis mice. Bioinformatics analysis revealed that IPF patients with higher TREM2 expression had a shorter survival time, and that TREM2 expression was closely associated with fibroblasts and M2 macrophages. Gene Ontology (GO) enrichment analysis showed that found TREM2-related differentially expressed genes (DEGs) were associated with inflammatory responses, extracellular matrix (ECM) and collagen formation. Single-cell RNA sequencing analysis revealed that TREM2 was predominantly expressed in macrophages. TREM2 insufficiency inhibited BLM-induced pulmonary fibrosis and M2 macrophage polarization. Mechanistic studies showed that TREM2 insufficiency suppressed the activation of STAT6 and the expression of fibrotic factors such as Fibronectin (Fib), Collagen I (Col I) and α- smooth muscle actin (α-SMA). CONCLUSION: Our study showed that TREM2 insufficiency might alleviate pulmonary fibrosis possibly through macrophage polarization regulation via STAT6 activation, providing a promising macrophage-related approach for the clinical therapy of pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , Pulmón , Ratones , Animales , Pulmón/patología , Fibrosis Pulmonar Idiopática/genética , Bleomicina/metabolismo , Macrófagos/metabolismo , Colágeno Tipo I/metabolismo , Ratones Endogámicos C57BL , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
12.
Mater Horiz ; 10(4): 1342-1353, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36723012

RESUMEN

Engineering chem-/sono-/photo-multimodal antitumor therapies has become an efficient strategy to combat malignant tumors. However, the existence of hypoxia in the tumor microenvironment (TME) leads to limited sonodynamic or photodynamic efficiency because O2 is the key reactant during the process of generation of reactive oxygen species (ROS). Here, to design a desirable platform that can simultaneously convert H2O2 in the TME into ROS and O2 for efficient chem-/sono-/photo-multimodal tumor therapies, we have created ultrasmall Cu2O-coordinated carbon nitride on a biocompatible ceria substrate (denoted as Cu2O-CNx@CeO2) via a self-assisted catalytic growth strategy. The chemical and morphological structures, ROS and O2 generation activities, and chemo-/photo-/sono-dynamic specificities of Cu2O-CNx@CeO2 when serving as multifunctional biocatalytic agents were systematically disclosed. The experimental studies validated that Cu2O-CNx@CeO2 presents state-of-the-art peroxidase-like and catalase-like activities. Moreover, the light excitation and ultrasound irradiation were also demonstrated to boost ROS production. The in vitro and in vivo experiments suggest that Cu2O-CNx@CeO2 can efficiently inhibit the growth of malignant melanoma via chem-/sono-/photo-multimodal antitumor ability. We believe that applying these new biocatalysts with dual catalytic activities of producing ROS and O2 will offer a new path for engineering multimodal nanoagents to combat malignant tumors.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Humanos , Especies Reactivas de Oxígeno , Terapia Combinada , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
13.
Arch Med Res ; 54(2): 124-134, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36759293

RESUMEN

BACKGROUND: Gastric cancer (GC) is often diagnosed at an advanced stage and thus patients have a poor prognosis. This implies that early detection of this cancer will improve patient prognosis and survival. This systematic review explored the association of circulating protein and metabolite biomarkers with GC development. METHODS: A literature search was conducted until November 2021 on Medline, Embase, Cochrane library, and Web of Science databases. Studies were included if they assessed circulating proteins and metabolites in blood, urine, or saliva and determined their association with GC risk. Quality of identified studies was determined using the Newcastle-Ottawa scale for cohort studies. Random and fixed effects meta-analyses were performed to calculate pooled odds ratio. RESULTS: A total of 53 studies were included. High levels of anti-Helicobacter pylORi IgG levels, pepsinogen I (PGI) <30 µg/L and serum pepsinogen I/ pepsinogen II (PGI/II) ratio<3 were positively associated with risk of developing GC (pooled odds ratio (OR): 2.70; 95% CI: 1.44-5.04, 5.96, 95% CI: 2.65-13.42 and 4.43; 95% CI: 3.04-6.47). In addition, an inverse relationship was found between ferritin, iron and transferrin levels and risk of developing GC (OR: 0.62; 95% CI: 0.38-1,0.97; 95% CI: 0.94-1 and 0.85; 95% CI: 0.76-0.94). However, there was no association between levels of glucose, cholesterol, vitamin C, vitamin B12, vitamin A, α-Carotene, ß-Carotene, α-Tocopherol, γ-Tocopherol, and GC risk. CONCLUSION: The pooled analysis demonstrated that high levels of anti-Helicobacter pylORi IgG, PGI<30µg/L and serum PGI/II ratio <3 and low levels of ferritin, iron and transferrin were associated with risk of GC.


Asunto(s)
Infecciones por Helicobacter , Neoplasias Gástricas , Humanos , Pepsinógeno A , Biomarcadores , Pepsinógeno C , Inmunoglobulina G , Ferritinas , Hierro , Transferrinas
14.
Nanomedicine ; 49: 102660, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36746273

RESUMEN

The synergistic effect of chemotherapy and photo-dynamic therapy (PDT) is an effective way to improve the efficiency of tumor treatment. However, most synergistic therapeutic drugs have poor water solubility and stability, so it is difficult to achieve high therapeutic effects while avoiding the severe side effects. Herein, a unique dandelion-like nanomedicine (named as cRGDfk-CCPT-mCe6) was successfully synthesized using Ce6-loaded amphiphilic ß-cyclodextrins (ß-CD) doped lipid-based vesicles as the core (receptacle) and ß-CD modified camptothecin (CPT) pro-drug as the flyable dandelion seeds. The ß-CD modified CPT pro-drug was introduced into the core vesicles in succession via host-guest interaction between inter-molecular ß-CD and CPT, and cRGDfk peptides were further introduced as the outermost layer (stigma) to enhance the internalization into cancer cells. CPT interacted with ß-CD through glutathione (GSH)-cleavable disulfide bonds, which led to drug release in glutathione-rich cancer cells, just as spread of dandelion seeds in the wind. GSH consumption further disrupted the intracellular redox homeostasis of cancer cells through combined action of Ce6 with light irradiation and the synergistic anti-tumor effect was thus achieved, resulting in apoptosis of cancer cells. Therefore, the nanomedicine provides a facile and versatile anti-tumor strategy, as well as a persistent anti-cancer effects.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Humanos , Profármacos/química , Camptotecina/farmacología , Camptotecina/uso terapéutico , Camptotecina/química , Nanomedicina , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Glutatión , Línea Celular Tumoral
15.
Front Endocrinol (Lausanne) ; 13: 1039786, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36465663

RESUMEN

Gastric cancer (GC) is a common lethal malignancy worldwide. Gastroscopy is an effective screening technique for decreasing mortality. However, there are still limited useful non-invasive markers for early detection of GC. Bile acids are important molecules for the modulation of energy metabolism. With an in-depth targeted method for accurate quantitation of 80 bile acids (BAs), we aimed to find potential biomarkers for the early screening of GC. A cohort with 280 participants was enrolled, including 113 GC, 22 benign gastric lesions (BGL) and 145 healthy controls. Potential markers were identified using a random forest machine algorithm in the discovery cohort (n=180), then validated in an internal validation cohort (n=78) and a group with 22 BGL. The results represented significant alterations in the circulating BA pool between GC and the controls. BAs also exhibited significant correlations with various clinical traits. Then, we developed a diagnostic panel that comprised six BAs or ratios for GC detection. The panel showed high accuracy for the diagnosis of GC with AUC of 1 (95%CI: 1.00-1.00) and 0.98 (95%CI: 0.93-1.00) in the discovery and validation cohort, respectively. This 6-BAs panel was also able to identify early GC with AUC of 1 (95%CI: 0.999-1.00) and 0.94 (95%CI: 0.83-1.00) in the discovery and validation cohort, respectively. Meanwhile, this panel achieved a good differential diagnosis between GC and BGL and the AUC was 0.873 (95%CI: 0.812-0.934). The alternations of serum bile acids are characteristic metabolic features of GC. Bile acids could be promising biomarkers for the early diagnosis of GC.


Asunto(s)
Ácidos y Sales Biliares , Neoplasias Gástricas , Humanos , Estudios de Casos y Controles , Neoplasias Gástricas/diagnóstico , Metabolómica , Biomarcadores
16.
Front Immunol ; 13: 951137, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35990657

RESUMEN

Background: Currently available prognostic tools and focused therapeutic methods result in unsatisfactory treatment of gastric cancer (GC). A deeper understanding of human epidermal growth factor receptor 2 (HER2)-coexpressed metabolic pathways may offer novel insights into tumour-intrinsic precision medicine. Methods: The integrated multi-omics strategies (including transcriptomics, proteomics and metabolomics) were applied to develop a novel metabolic classifier for gastric cancer. We integrated TCGA-STAD cohort (375 GC samples and 56753 genes) and TCPA-STAD cohort (392 GC samples and 218 proteins), and rated them as transcriptomics and proteomics data, resepectively. 224 matched blood samples of GC patients and healthy individuals were collected to carry out untargeted metabolomics analysis. Results: In this study, pan-cancer analysis highlighted the crucial role of ERBB2 in the immune microenvironment and metabolic remodelling. In addition, the metabolic landscape of GC indicated that alanine, aspartate and glutamate (AAG) metabolism was significantly associated with the prevalence and progression of GC. Weighted metabolite correlation network analysis revealed that glycolysis/gluconeogenesis (GG) and AAG metabolism served as HER2-coexpressed metabolic pathways. Consensus clustering was used to stratify patients with GC into four subtypes with different metabolic characteristics (i.e. quiescent, GG, AAG and mixed subtypes). The GG subtype was characterised by a lower level of ERBB2 expression, a higher proportion of the inflammatory phenotype and the worst prognosis. However, contradictory features were found in the mixed subtype with the best prognosis. The GG and mixed subtypes were found to be highly sensitive to chemotherapy, whereas the quiescent and AAG subtypes were more likely to benefit from immunotherapy. Conclusions: Transcriptomic and proteomic analyses highlighted the close association of HER-2 level with the immune status and metabolic features of patients with GC. Metabolomics analysis highlighted the co-expressed relationship between alanine, aspartate and glutamate and glycolysis/gluconeogenesis metabolisms and HER2 level in GC. The novel integrated multi-omics strategy used in this study may facilitate the development of a more tailored approach to GC therapy.


Asunto(s)
Neoplasias Gástricas , Alanina , Ácido Aspártico/genética , Glutamatos/genética , Humanos , Inmunoterapia , Metabolómica , Terapia Neoadyuvante , Proteómica , Receptor ErbB-2 , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Transcriptoma , Microambiente Tumoral/genética
17.
Sci Rep ; 11(1): 23649, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34880328

RESUMEN

Intrahepatic cholangiocarcinoma (CHOL) remains a rare malignancy, ranking as the leading lethal primary liver cancer worldwide. However, the biological functions of integrator complex subunit 8 (INTS8) in CHOL remain unknown. Thus, this research aimed to explore the potential role of INTS8 as a novel diagnostic or therapeutic target in CHOL. Differentially expressed genes (DEGs) in two Gene Expression Omnibus (GEO) datasets were obtained by the "RRA" package in R software. The "maftools" package was used to visualize the CHOL mutation data from The Cancer Genome Atlas (TCGA) database. The expression of INTS8 was detected by performing quantitative reverse transcription-PCR (qRT-PCR) and immunohistochemistry in cell lines and human samples. The association between subtypes of tumour-infiltrating immune cells (TIICs) and INTS8 expression in CHOL was determined by using CIBERSORT tools. We evaluated the correlations between INTS8 expression and mismatch repair (MMR) genes and DNA methyltransferases (DNMTs) in pan-cancer analysis. Finally, the pan-cancer prognostic signature of INTS8 was identified by univariate analysis. We obtained the mutation landscapes of an RRA gene set in CHOL. The expression of INTS8 was upregulated in CHOL cell lines and human CHOL samples. Furthermore, INTS8 expression was closely associated with a distinct landscape of TIICs, MMR genes, and DNMTs in CHOL. In addition, the high INTS8 expression group presented significantly poor outcomes, including overall survival (OS), disease-specific survival (DSS) and disease-free interval (DFI) (p < 0.05) in pan-cancer. INTS8 contributes to the tumorigenesis and progression of CHOL. Our study highlights the significant role of INTS8 in CHOL and pan-cancers, providing a valuable molecular target for cancer research.


Asunto(s)
Neoplasias de los Conductos Biliares/terapia , Colangiocarcinoma/terapia , Biología Computacional/métodos , Subunidades de Proteína/fisiología , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , Subunidades de Proteína/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
ACS Appl Mater Interfaces ; 13(42): 49631-49641, 2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34636534

RESUMEN

Bioinspired strategies have recently emerged as novel approaches for designing a functionalized nanovector with enhanced tumor targeting and therapeutic efficacy. Herein, a virus-like Fe3O4/Au@C nanovector is described for programmable drug delivery via hierarchical targeting. Specifically, the virus-like Fe3O4/Au@C nanovector is synthesized via a simple hydrothermal process, and then the spiky surface of which is camouflaged via doxorubicin (DOX)-conjugated polyethylene glycol (PEG), constructing an innovative virus-like core/spherical shell biomimetic nanomedicine (Fe3O4/Au@C-DOX-PEG), which is conducive to improve bioavailability and reduce adverse effects. After systemic administration, the as-prepared nanomedicine is capable of facilitating effective tumor accumulation and deep tumor penetration with the assistance of an external magnetic field and endogenous pH stimuli. Simultaneously, in response to the acidic tumor microenvironment, Fe3O4/Au@C-DOX nanocomposites are released and exhibit excellent performance in cellular internalization through a virus-mimetic rough surface. Furthermore, the in vivo experiments identify that the unique nanomedicine is bestowed with an effective targeting tumor, prominent antitumor efficacy, and reduced systemic toxicity. Such a bioinspired hierarchical targeting nanoplatform holds promising potential for enhanced chemotherapeutic intracellular delivery and tumor theranostics.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Materiales Biomiméticos/farmacología , Carbono/química , Doxorrubicina/farmacología , Oro/química , Nanopartículas de Magnetita/química , Nanopartículas/química , Animales , Antibióticos Antineoplásicos/química , Materiales Biomiméticos/química , Proliferación Celular/efectos de los fármacos , Doxorrubicina/química , Portadores de Fármacos/química , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Células MCF-7 , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ensayo de Materiales , Ratones , Nanomedicina , Polietilenglicoles/química , Polietilenglicoles/farmacología
19.
Front Genet ; 12: 692953, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34490033

RESUMEN

Background: Pancreatic adenocarcinoma (PAAD) has a considerably bad prognosis, and its pathophysiologic mechanism remains unclear. Thus, we aimed to identify real hub genes to better explore the pathophysiology of PAAD and construct a prognostic panel to better predict the prognosis of PAAD using the weighted gene co-expression network analysis (WGCNA) and the least absolute shrinkage and selection operator (LASSO) algorithms. Methods: WGCNA identified the modules most closely related to the PAAD stage and grade based on the Gene Expression Omnibus. The module genes significantly associated with PAAD progression and prognosis were considered as the real hub genes. Eligible genes in the most significant module were selected for construction and validation of a multigene prognostic signature based on the LASSO-Cox regression analysis in The Cancer Genome Atlas and the International Cancer Genome Consortium databases, respectively. Results: The brown module identified by WGCNA was most closely associated with the clinical characteristics of PAAD. Scaffold attachment factor B (SAFB) was significantly associated with PAAD progression and prognosis, and was identified as the real hub gene of PAAD. Moreover, both transcriptional and translational levels of SAFB were significantly lower in PAAD tissues compared with normal pancreatic tissues. In addition, a novel multigene-independent prognostic signature consisting of SAFB, SP1, and SERTAD3 was identified and verified. The predictive accuracy of our signature was superior to that of previous studies, especially for predicting 3- and 5-year survival probabilities. Furthermore, a prognostic nomogram based on independent prognostic variables was developed and validated using calibration curves. The predictive ability of this nomogram was also superior to the well-established AJCC stage and histological grade. The potential mechanisms of different prognoses between the high- and low-risk subgroups were also investigated using functional enrichment analysis, GSEA, ssGSEA, immune checkpoint analysis, and mutation profile analysis. Conclusion: SAFB was identified as the real hub gene of PAAD. A novel multigene-independent prognostic signature was successfully identified and validated to better predict PAAD prognosis. An accurate nomogram was also developed and verified to aid in the accurate treatment of tumors, as well as in early intervention.

20.
Biomater Sci ; 9(16): 5542-5550, 2021 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-34254095

RESUMEN

Multi-functionality has been a constant pursuit in the development of next-generation drug carriers, as it will bring the potential for combination therapy by integrating diverse therapeutic modes. In this work, FeSe2 nanosheets (NSs) have been prepared as a bifunctional platform to investigate their use in synergistic cancer therapy. Bifunctional FeSe2 NSs exhibit exceptional Fenton-like activity that generates cytotoxic hydroxyl radical (˙OH) and strong broad photothermal performance including the second-infrared (NIR-II) spectral range, wherein the ˙OH production can be enhanced by NIR-II light irradiation. Furthermore, doxorubicin (DOX) was conjugated onto NSs via a pH-responsive hydrazone bond to achieve preferential drug release in an acidic microenvironment. Upon intratumoral administration, these bifunctional drug-carrying FeSe2 NSs showed an NIR-II irradiation-reinforced strong tumor suppression effect, and no obvious toxicity to normal tissues was observed. This study provides a new paradigm for the design of advanced drug carriers relying on their inherent physicochemical properties.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Línea Celular Tumoral , Doxorrubicina , Liberación de Fármacos , Humanos , Neoplasias/tratamiento farmacológico , Fototerapia , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA