Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 16(14): 18019-18029, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38546167

RESUMEN

With the continuous advancement of electrodialysis (ED) technology, there arises a demand for improved monovalent cation exchange membranes (CEMs). However, limitations in membrane materials and structures have resulted in the low selectivity of monovalent CEMs, posing challenges in the separation of Li+ and Mg2+. In this investigation, a designed CEM with a swelling-embedded structure was created by integrating a polyelectrolyte containing N-oxide Zwitterion into a sulfonated poly(ether ether ketone) (SPEEK) membrane, leveraging the notable solubility characteristic of SPEEK. The membranes were prepared by using N-oxide zwitterionic polyethylenimine (ZPEI) and 1,3,5-benzenetrlcarbonyl trichloride (TMC). The as-prepared membranes underwent systematic characterization and testing, evaluating their structural, physicochemical, electrochemical, and selective ED properties. During ED, the modified membranes demonstrated notable permeability selectivity for Li+ ions in binary (Li+/Mg2+) systems. Notably, at a constant current density of 2.5 mA cm-2, the modified membrane PEI-TMC/SPEEK exhibited significant permeability selectivity (PMg2+Li+=5.63) in the Li+/Mg2+ system, while ZPEI-TMC/SPEEK outperformed, displaying remarkable permeability selectivity (PMg2+Li+=12.43) in the Li+/Mg2+ system, surpassing commercial monovalent cation-selective membrane commercial monovalent cation-selective membrane (CIMS). Furthermore, in the Li+/Mg2+ binary system, Li+ flux reached 9.78 × 10-9 mol cm-2 s-1 for ZPEI-TMC/SPEEK, while its Mg2+ flux only reached 2.7 × 10-9 mol cm-2 s-1, showing potential for lithium-magnesium separation. In addition, ZPEI-TMC/SPEEK was tested for performance and stability at high current densities. This work offers a straightforward preparation process and an innovative structural approach, presenting methodological insights for the advancement of lithium and magnesium separation techniques.

2.
Hepatology ; 79(3): 650-665, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37459556

RESUMEN

BACKGROUND AND AIMS: Hepatoblastoma (HB) is the most common liver cancer in children, posing a serious threat to children's health. Chemoresistance is the leading cause of mortality in patients with HB. A more explicit definition of the features of chemotherapy resistance in HB represents a fundamental urgent need. APPROACH AND RESULTS: We performed an integrative analysis including single-cell RNA sequencing, whole-exome sequencing, and bulk RNA sequencing in 180 HB samples, to reveal genomic features, transcriptomic profiles, and the immune microenvironment of HB. Multicolor immunohistochemistry staining and in vitro experiments were performed for validation. Here, we reported four HB transcriptional subtypes primarily defined by differential expression of transcription factors. Among them, the S2A subtype, characterized by strong expression of progenitor ( MYCN , MIXL1 ) and mesenchymal transcription factors ( TWIST1 , TBX5 ), was defined as a new chemoresistant subtype. The S2A subtype showed increased TGF-ß cancer-associated fibroblast and an immunosuppressive microenvironment induced by the upregulated TGF-ß of HB. Interestingly, the S2A subtype enriched SBS24 signature and significantly higher serum aflatoxin B1-albumin (AFB1-ALB) level in comparison with other subtypes. Functional assays indicated that aflatoxin promotes HB to upregulate TGF-ß. Furthermore, clinical prognostic analysis showed that serum AFB1-ALB is a potential indicator of HB chemoresistance and prognosis. CONCLUSIONS: Our studies offer new insights into the relationship between aflatoxin and HB chemoresistance and provide important implications for its diagnosis and treatment.


Asunto(s)
Aflatoxinas , Hepatoblastoma , Neoplasias Hepáticas , Niño , Humanos , Hepatoblastoma/genética , Hepatoblastoma/metabolismo , Factor de Crecimiento Transformador beta , Neoplasias Hepáticas/metabolismo , Factores de Transcripción/genética , Fenotipo , Microambiente Tumoral
3.
Small ; 20(12): e2306313, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37948422

RESUMEN

Ion resource recovery from organic wastewater is beneficial for achieving emission peaks and carbon neutrality targets. Advanced organic solvent-resistant anion exchange membranes (AEMs) for treating organic wastewater via electrodialysis (ED) are of significant interest. Herein, a kind of 3D network AEM based on poly(arylene ether sulfone) cross-linked with a flexible cross-linker (DBH) for ion resource recovery via ED in organic solvent system is reported. Investigations demonstrate that the as-prepared AEMs show excellent dimensional stability in 60% DMSO (aq.), 60% ethanol (aq.), and 60% acetone (aq.), respectively. For example, the optimized AEM shows very low swelling ratios of 1.04-1.10% in the organic solvents. ED desalination ratio can reach 99.1% after exposure of the AEM to organic solvents for 30 days, and remain > 99% in a mixture solution containing organic solvents and 0.5 m NaCl. Additionally, at a current density of 2.5 mA cm-2, the optimized AEM soaked in organic solvents for 30 days shows a high perm-selectivity (Cl-/SO4 2-) of 133.09 (vs 13.11, Neosepta ACS). The superior ED performance is attributed to the stable continuous sub-nanochannels within AEM confirmed by SAXS, rotational energy barriers, etc. This work shows the potential application of cross-linked AEMs for resource recovery in organic wastewater.

4.
Hepatology ; 79(4): 780-797, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37725755

RESUMEN

BACKGROUND AND AIMS: Scirrhous HCC (SHCC) is one of the unique subtypes of HCC, characterized by abundant fibrous stroma in the tumor microenvironment. However, the molecular traits of SHCC remain unclear, which is essential to develop specialized therapeutic approaches for SHCC. APPROACH AND RESULTS: We presented an integrative analysis containing single-cell RNA-sequencing, whole-exome sequencing, and bulk RNA-sequencing in SHCC and usual HCC samples from 134 patients to delineate genomic features, transcriptomic profiles, and stromal immune microenvironment of SHCC. Multiplexed immunofluorescence staining, flow cytometry, and functional experiments were performed for validation. Here, we identified SHCC presented with less genomic heterogeneity while possessing a unique transcriptomic profile different from usual HCC. Insulin-like growth factor 2 was significantly upregulated in SHCC tumor cells compared to usual HCC, and could serve as a potential diagnostic biomarker for SHCC. Significant tumor stromal remodeling and hypoxia were observed in SHCC with enrichment of matrix cancer-associated fibroblasts and upregulation of hypoxic pathways. Insulin-like growth factor 2 was identified as a key mediator in shaping the hypoxic stromal microenvironment of SHCC. Under this microenvironment, SHCC exhibited an immunosuppressive niche correlated to enhanced VEGFA signaling activity, where CD4 + T cells and CD8 + T cells were dysfunctional. Furthermore, we found that another hypoxic-related molecule SPP1 from SHCC tumor cells suppressed the function of dendritic cells via the SPP1-CD44 axis, which also probably hindered the activation of T cells. CONCLUSION: We uncovered the genomic characteristics of SHCC, and revealed a hypoxia-driven tumor stroma remodeling and immunosuppressive microenvironment in SHCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Hipoxia/metabolismo , Transducción de Señal , ARN , Microambiente Tumoral
5.
ACS Nano ; 17(23): 23405-23421, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37988576

RESUMEN

Radiotherapy causes DNA damage by direct ionization and indirect generation of reactive oxygen species (ROS) thereby destroying cancer cells. However, ionizing radiation (IR) unexpectedly elicits metastasis and invasion of cancer cells by inducing cancer stem cells' (CSCs) properties. As BMI1 is a crucial gene that causes radioresistance and an unfavorable prognosis of hepatocellular carcinoma (HCC), BMI1 inhibitor PTC-209 has been encapsulated in a ROS-responsive liposome (LP(PTC-209)) to be temporally and spatially delivered to radioresistant HCC tissue. The ROS generated during IR was not only considered to directly cause tumor cell death but also be used as a stimulator to trigger ROS-responsive drug release from LP(PTC-209). The PTC-209 released into resistant HCC tissue under radiotherapy further led to cancer stem cell (CSC) differentiation and then recovered radiosensitivity of HCC tumor. The suppression of the radioresistant performance of LP(PTC-209) has been proved on radiosensitive and radioresistant Hepa1-6 CSC tumor models, respectively. Our study clarified the relationship between radiotherapy and cancer stemness and provided insights to achieve complete suppression of radioresistant HCC tumor by inhibiting cancer stemness.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/radioterapia , Liposomas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/radioterapia , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Tolerancia a Radiación , Complejo Represivo Polycomb 1/metabolismo
6.
Gastroenterol Rep (Oxf) ; 11: goad060, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37842201

RESUMEN

Background: Insufficient post-operative future liver remnant (FLR) limits the feasibility of hepatectomy for patients. Staged hepatectomy is an effective surgical approach that can improve the resection rate of hepatocellular carcinoma (HCC). This study aimed to compare the safety and efficacy of laparoscopic microwave ablation and portal vein ligation for staged hepatectomy (LAPS) and classical associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) in the treatment of hepatitis B virus (HBV)-related HCC. Methods: Clinical data of patients with HBV-related HCC who underwent LAPS or ALPPS in our institute between January 2017 and May 2022 were retrospectively analysed. Results: A total of 18 patients with HBV-related HCC were retrospectively analysed and divided into the LAPS group (n = 9) and ALPPS group (n = 9). Eight patients in the LAPS group and eight patients in the ALPPS group proceeded to a similar resection rate (88.9% vs 88.9%, P = 1.000). The patients undergoing LAPS had a lower total comprehensive complication index than those undergoing ALPPS but there was not a significant different between the two groups (8.66 vs 35.87, P = 0.054). The hypertrophy rate of FLR induced by ALPPS tended to be more rapid than that induced by LAPS (24.29 vs 13.17 mL/d, P = 0.095). The 2-year recurrence-free survival (RFS) was 0% for ALPPS and 35.7% for LAPS (P = 0.009), whereas the 2-year overall survival for ALPPS and LAPS was 33.3% and 100.0% (P = 0.052), respectively. Conclusions: LAPS tended to induce lower morbidity and FLR hypertrophy more slowly than ALPPS, with a comparable resection rate and better long-term RFS in HBV-related HCC patients.

7.
Adv Sci (Weinh) ; 10(30): e2303588, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37697634

RESUMEN

Constructing a functional layer on the surface of commercial membrane (as a substrate) to inhibit the formation of biofilms is an efficient strategy to prepare an antibacterial anion exchange membrane (AEM). Herein, a rechargeable multifunctional anti-biological system is reported by utilizing the mussel-inspired L-dopa connection function on commercial AEMs. Cobalt nanoparticles (Co NPs) and N-chloramine compounds are deposited on the AEM surface by a two-step modification procedure. The anti-biofouling abilities of the membranes are qualitatively and quantitatively analyzed by adopting common Gram-negative (E. coli) and Gram-positive (S. aureus & Bacillus) bacteria as model biofouling organisms. The optimized membrane exhibits a high stability concerning the NaCl solution separation performance within 240 min. Meantime, the mechanism of the anti-adhesion is un-veiled at an atomic level and molecular dynamics (MD) simulation are conducted to measure the interaction, adsorption energy and average loading by using lipopolysaccharide (LPS) of E. coli. In view of the superior performance of antibacterial surfaces, it is believed that this work could provide a valuable guideline for the design of membrane materials with resistance to biological contamination.


Asunto(s)
Escherichia coli , Staphylococcus aureus , Bacterias , Antibacterianos/farmacología , Antibacterianos/química
8.
Mol Ther ; 31(11): 3225-3242, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37735874

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is a deadly cancer with rapid tumor progression. While hyperactive mRNA translation caused by mis-regulated mRNA or tRNA modifications promotes ICC development, the role of rRNA modifications remains elusive. Here, we found that 18S rRNA m6A modification and its methyltransferase METTL5 were aberrantly upregulated in ICC and associated with poorer survival (log rank test, p < 0.05). We further revealed the critical role of METTL5-mediated 18S rRNA m6A modification in regulation of ICC cell growth and metastasis using loss- and gain-of function assays in vitro and in vivo. The oncogenic function of METTL5 is corroborated using liver-specific knockout and overexpression ICC mouse models. Mechanistically, METTL5 depletion impairs 18S rRNA m6A modification that hampers ribosome synthesis and inhibits translation of G-quadruplex-containing mRNAs that are enriched in the transforming growth factor (TGF)-ß pathway. Our study uncovers the important role of METTL5-mediated 18S rRNA m6A modification in ICC and unravels the mechanism of rRNA m6A modification-mediated oncogenic mRNA translation control.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Animales , Ratones , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , Colangiocarcinoma/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Biosíntesis de Proteínas , Línea Celular Tumoral
9.
Adv Healthc Mater ; 12(30): e2302013, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37665720

RESUMEN

Radiofrequency ablation (RFA) is a widely used therapy for hepatocellular carcinoma (HCC). However, in cases of insufficient RFA (iRFA), nonlethal temperatures in the transition zone increase the risk of postoperative relapse. The pathological analysis of HCC tissues shows that iRFA-induced upregulation of myeloid-derived suppressor cells (MDSCs) in residual tumors is critical for postoperative recurrence. Furthermore, this study demonstrates, for the first time, that combining MDSCs suppression strategy during iRFA can unexpectedly lead to a compensatory increase in PD-L1 expression on the residual MDSCs, attributed to relapse due to immune evasion. To address this issue, a novel size-tunable hybrid nano-microliposome is designed to co-deliver MDSCs inhibitors (IPI549) and αPDL1 antibodies (LPIP) for multipathway activation of immune responses. The LPIP is triggered to release immune regulators by the mild heat in the transition zone of iRFA, selectively inhibiting MDSCs and blocking the compensatory upregulation of PD-L1 on surviving MDSCs. The combined strategy of LPIP + iRFA effectively ablates the primary tumor by activating immune responses in the transition zone while suppressing the compensatory immune evasion of surviving MDSCs. This approach avoids the relapse of the residual tumor in a post-iRFA incomplete ablation model and appears to be a promising strategy in RFA for the eradication of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Células Supresoras de Origen Mieloide , Ablación por Radiofrecuencia , Humanos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Antígeno B7-H1 , Evasión Inmune , Recurrencia Local de Neoplasia , Recurrencia
10.
J Cancer Res Clin Oncol ; 149(13): 12365-12377, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37436513

RESUMEN

BACKGROUND: Liver resection is the mainstay of curative treatment for intrahepatic cholangiocarcinoma (ICC) while the postoperative prognosis varies greatly, with no recognized biomarker. We aimed to identify the plasma metabolomic biomarkers that could be used for preoperative risk stratification of ICC patients. METHODS: 108 eligible ICC patients who underwent radical surgical resection between August 2012 and October 2020 were enrolled. Patients were randomly divided into a discovery cohort (n = 76) and a validation cohort (n = 32) by 7:3. Metabolomics profiling of preoperative plasma was performed and clinical data were collected. The least absolute shrinkage and selection operator (LASSO) regression, Cox regression, and receiver operating characteristic (ROC) analyses were used to screen and validate the survival-related metabolic biomarker panel and construct a LASSO-Cox prediction model. RESULTS: 10 survival-related metabolic biomarkers were used for construction of a LASSO-Cox prediction model. In the discovery and validation cohorts, the LASSO-Cox prediction model achieved an AUC of 0.876 (95%CI: 0.777-0.974) and 0.860 (95%CI: 0.711-1.000) in evaluating 1-year OS of ICC patients, respectively. The OS of ICC patients in the high-risk group was significantly worse than that in the low-risk group (discovery cohort, p < 0.0001; validation cohort: p = 0.041). Also, the LASSO-Cox risk score (HR 2.43, 95%CI: 1.81-3.26, p < 0.0001) was a significant independent risk factor associated with OS. CONCLUSIONS: The LASSO-Cox prediction model has potential as an important tool in evaluating the OS of ICC patients after surgical resection and can be used as prediction tools to implement the best treatment options that could result in better outcomes.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Colangiocarcinoma/cirugía , Colangiocarcinoma/patología , Pronóstico , Neoplasias de los Conductos Biliares/cirugía , Conductos Biliares Intrahepáticos , Medición de Riesgo
11.
Hepatology ; 77(6): 1896-1910, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35698894

RESUMEN

BACKGROUND AND AIMS: Radiotherapy is an increasingly essential therapeutic strategy in the management of hepatocellular carcinoma (HCC). Nevertheless, resistance to radiotherapy is one of the primary obstacles to successful treatment outcomes. Hence, we aim to elucidate the mechanisms underlying radioresistance and identify reliable biotargets that would be inhibited to enhance the efficacy of radiotherapy in HCC. APPROACH AND RESULTS: From a label-free quantitative proteome screening, we identified transfer RNA (tRNA; guanine- N [7]-) methyltransferase 1 (METTL1), a key enzyme for N7-methylguanosine (m 7 G) tRNA modification, as an essential driver for HCC cells radioresistance. We reveal that METTL1 promotes DNA double-strand break (DSB) repair and renders HCC cells resistant to ionizing radiation (IR) using loss-of-function and gain-of-function assays in vitro and in vivo. Mechanistically, METTL1-mediated m 7 G tRNA modification selectively regulates the translation of DNA-dependent protein kinase catalytic subunit or DNA ligase IV with higher frequencies of m 7 G-related codons after IR treatment, thereby resulting in the enhancement of nonhomologous end-joining (NHEJ)-mediated DNA DSB repair efficiency. Clinically, high METTL1 expression in tumor tissue is significantly correlated with poor prognosis in radiotherapy-treated patients with HCC. CONCLUSIONS: Our findings show that METTL1 is a critical enhancer for HCC cell NHEJ-based DNA repair following IR therapy. These findings give insight into the role of tRNA modification in messenger RNA translation control in HCC radioresistance.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Reparación del ADN , Metiltransferasas/genética , ARN de Transferencia
12.
Cancer Res ; 83(1): 89-102, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36102722

RESUMEN

The tyrosine kinase inhibitor lenvatinib is a first-line drug for treating patients with advanced hepatocellular carcinoma (HCC). However, its efficacy is severely hampered by drug resistance. Insights into the molecular mechanisms underlying lenvatinib resistance could provide new strategies to improve and prolong responses. Here, we performed unbiased proteomic screening of parental and lenvatinib-resistant HCC cells and discovered that methyltransferase-like protein-1 (METTL1) and WD repeat domain 4 protein (WDR4), the two key components of the tRNA N7-methylguanosine (m7G) methyltransferase complex, were dramatically upregulated in lenvatinib-resistant cells. METTL1 knockdown overrode resistance by impairing the proliferation capacity of HCC cells and promoting apoptosis under lenvatinib treatment. In addition, overexpression of wild-type METTL1 but not its catalytic dead mutant induced lenvatinib resistance. Animal experiments including hydrodynamic injection, subcutaneous implantation, and orthotopic xenograft mouse models further demonstrated the critical function of METTL1/WDR4-mediated m7G tRNA modification in promoting lenvatinib resistance in vivo. Mechanistically, METTL1 promoted translation of EGFR pathway genes to trigger drug resistance. This work reveals the important role of METTL1-mediated m7G tRNA modification in promoting lenvatinib resistance and provides a promising prediction marker and intervention target for resistance. SIGNIFICANCE: Upregulation of tRNA m7G methyltransferase complex components METTL1 and WDR4 promotes lenvatinib resistance in HCC and confers a sensitivity to METTL1 targeting, providing a promising strategy to override resistance.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animales , Ratones , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteómica , ARN de Transferencia/genética , Proteínas de Unión al GTP/metabolismo
13.
ACS Nano ; 16(3): 4629-4641, 2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35226457

RESUMEN

Ion-conductive polymers having a well-defined phase-separated structure show the potential application of separating mono- and bivalent ion separation. In this work, three side-chain-type poly(arylene ether sulfone)-based anion exchange membranes (AEMs) have been fabricated to investigate the effect of the stiffness of the polymer backbone within AEMs on the Cl-/NO3- and Cl-/SO42- separation performance. Our investigations via small-angle X-ray scattering (SAXS), positron annihilation, and differential scanning calorimetry (DSC) demonstrate that the as-prepared AEM with a rigid benzimidazole structure in the backbone bears subnanometer ion channels resulting from the arrangement of the rigid polymer backbone. In particular, SAXS results demonstrate that the rigid benzimidazole-containing AEM in the wet state has an ion cluster size of 0.548 nm, which is smaller than that of an AEM with alkyl segments in the backbone (0.760 nm). Thus, in the electrodialysis (ED) process, the former exhibits a superior capacity of separating Cl-/SO42- ions relative to latter. Nevertheless, the benzimidazole-containing AEM shows an inability to separate the Cl-/NO3- ions, which is possibly due to the similar ion size of the two. The higher rotational energy barrier (4.3 × 10-3 Hartree) of benzimidazole units and the smaller polymer matrix free-volume (0.636%) in the AEM significantly contribute to the construction of smaller ion channels. As a result, it is believed that the rigid benzimidazole structure of this kind is a benefit to the construction of stable subnanometer ion channels in the AEM that can selectively separate ions with different sizes.

14.
Clin Transl Med ; 11(12): e661, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34898034

RESUMEN

BACKGROUND: N7 -methylguanosine (m7 G) modification is one of the most common transfer RNA (tRNA) modifications in humans. The precise function and molecular mechanism of m7 G tRNA modification in hepatocellular carcinoma (HCC) remain poorly understood. METHODS: The prognostic value and expression level of m7 G tRNA methyltransferase complex components methyltransferase-like protein-1 (METTL1) and WD repeat domain 4 (WDR4) in HCC were evaluated using clinical samples and TCGA data. The biological functions and mechanisms of m7 G tRNA modification in HCC progression were studied in vitro and in vivo using cell culture, xenograft model, knockin and knockout mouse models. The m7 G reduction and cleavage sequencing (TRAC-seq), polysome profiling and polyribosome-associated mRNA sequencing methods were used to study the levels of m7 G tRNA modification, tRNA expression and mRNA translation efficiency. RESULTS: The levels of METTL1 and WDR4 are elevated in HCC and associated with advanced tumour stages and poor patient survival. Functionally, silencing METTL1 or WDR4 inhibits HCC cell proliferation, migration and invasion, while forced expression of wild-type METTL1 but not its catalytic dead mutant promotes HCC progression. Knockdown of METTL1 reduces m7 G tRNA modification and decreases m7 G-modified tRNA expression in HCC cells. Mechanistically, METTL1-mediated tRNA m7 G modification promotes the translation of target mRNAs with higher frequencies of m7 G-related codons. Furthermore, in vivo studies with Mettl1 knockin and conditional knockout mice reveal the essential physiological function of Mettl1 in hepatocarcinogenesis using hydrodynamics transfection HCC model. CONCLUSIONS: Our work reveals new insights into the role of the misregulated tRNA modifications in liver cancer and provides molecular basis for HCC diagnosis and treatment.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/genética , Metiltransferasas/efectos adversos , Pronóstico , ARN de Transferencia/efectos de los fármacos , Animales , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/etiología , Modelos Animales de Enfermedad , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Noqueados
15.
Mol Cell ; 81(16): 3339-3355.e8, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34352206

RESUMEN

Cancer cells selectively promote translation of specific oncogenic transcripts to facilitate cancer survival and progression, but the underlying mechanisms are poorly understood. Here, we find that N7-methylguanosine (m7G) tRNA modification and its methyltransferase complex components, METTL1 and WDR4, are significantly upregulated in intrahepatic cholangiocarcinoma (ICC) and associated with poor prognosis. We further reveal the critical role of METTL1/WDR4 in promoting ICC cell survival and progression using loss- and gain-of-function assays in vitro and in vivo. Mechanistically, m7G tRNA modification selectively regulates the translation of oncogenic transcripts, including cell-cycle and epidermal growth factor receptor (EGFR) pathway genes, in m7G-tRNA-decoded codon-frequency-dependent mechanisms. Moreover, using overexpression and knockout mouse models, we demonstrate the crucial oncogenic function of Mettl1-mediated m7G tRNA modification in promoting ICC tumorigenesis and progression in vivo. Our study uncovers the important physiological function and mechanism of METTL1-mediated m7G tRNA modification in the regulation of oncogenic mRNA translation and cancer progression.


Asunto(s)
Colangiocarcinoma/genética , Proteínas de Unión al GTP/genética , Metiltransferasas/genética , Biosíntesis de Proteínas , Animales , Carcinogénesis/genética , Colangiocarcinoma/patología , Progresión de la Enfermedad , Receptores ErbB/genética , Guanosina/análogos & derivados , Guanosina/genética , Humanos , Ratones , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/genética , ARN de Transferencia/genética
16.
Membranes (Basel) ; 11(5)2021 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-34068766

RESUMEN

Developing monovalent cation permselective membranes (MCPMs) with high-efficient permselectivity is the core concern in specific industrial applications. In this work, we have fabricated a series of novel cation exchange membranes (CEMs) based on sulfonated polysulfone (SPSF) surface modification by polyethyleneimine (PEI) and 4'-aminobenzo-12-crown-4 (12C4) codeposited with dopamine (DA) successively, which was followed by the cross-linking of glutaraldehyde (GA). The as-prepared membranes before and after modification were systematically characterized with regard to their structures as well as their physicochemical and electrochemical properties. Particularly, the codeposition sequence of modified ingredients was investigated on galvanostatic permselectivity to cations. The modified membrane (M-12C4-0.50-PEI) exhibits significantly prominent selectivity to Li+ ions (PMg2+Li+ = 5.23) and K+ ions (PMg2+K+ = 13.56) in Li+/Mg2+ and K+/Mg2+ systems in electrodialysis (ED), which is far superior to the pristine membrane (M-0, PMg2+Li+ = 0.46, PMg2+K+ = 1.23) at a constant current density of 5.0 mA·cm-2. It possibly arises from the synergistic effects of electrostatic repulsion (positively charged PEI), pore-size sieving (distribution of modified ingredients), and specific interaction effect (12C4 ~Li+). This facile strategy may provide new insights into developing selective CEMs in the separation of specific cations by ED.

17.
Eur Radiol ; 31(11): 8615-8627, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33877387

RESUMEN

OBJECTIVES: Pretreatment evaluation of tumor biology and microenvironment is important to predict prognosis and plan treatment. We aimed to develop nomograms based on gadoxetic acid-enhanced MRI to predict microvascular invasion (MVI), tumor differentiation, and immunoscore. METHODS: This retrospective study included 273 patients with HCC who underwent preoperative gadoxetic acid-enhanced MRI. Patients were assigned to two groups: training (N = 191) and validation (N = 82). Univariable and multivariable logistic regression analyses were performed to investigate clinical variables and MRI features' associations with MVI, tumor differentiation, and immunoscore. Nomograms were developed based on features associated with these three histopathological features in the training cohort, then validated, and evaluated. RESULTS: Predictors of MVI included tumor size, rim enhancement, capsule, percent decrease in T1 images (T1D%), standard deviation of apparent diffusion coefficient, and alanine aminotransferase levels, while capsule, peritumoral enhancement, mean relaxation time on the hepatobiliary phase (T1E), and alpha-fetoprotein levels predicted tumor differentiation. Predictors of immunoscore included the radiologic score constructed by tumor number, intratumoral vessel, margin, capsule, rim enhancement, T1D%, relaxation time on plain scan (T1P), and alpha-fetoprotein and alanine aminotransferase levels. Three nomograms achieved good concordance indexes in predicting MVI (0.754, 0.746), tumor differentiation (0.758, 0.699), and immunoscore (0.737, 0.726) in the training and validation cohorts, respectively. CONCLUSION: MRI-based nomograms effectively predict tumor behaviors in HCC and may assist clinicians in prognosis prediction and pretreatment decisions. KEY POINTS: • This study developed and validated three nomograms based on gadoxetic acid-enhanced MRI to predict MVI, tumor differentiation, and immunoscore in patients with HCC. • The pretreatment prediction of tumor microenvironment may be useful to guide accurate prognosis and planning of surgical and immunological therapies for individual patients with HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/diagnóstico por imagen , Medios de Contraste , Gadolinio DTPA , Humanos , Neoplasias Hepáticas/diagnóstico por imagen , Imagen por Resonancia Magnética , Invasividad Neoplásica , Nomogramas , Estudios Retrospectivos , Microambiente Tumoral
18.
Hepatology ; 74(3): 1339-1356, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33638162

RESUMEN

BACKGROUND AND AIMS: The dynamic N6-methyladenosine (m6 A) mRNA modification is essential for acute stress response and cancer progression. Sublethal heat stress from insufficient radiofrequency ablation (IRFA) has been confirmed to promote HCC progression; however, whether m6 A machinery is involved in IRFA-induced HCC recurrence remains open for study. APPROACH AND RESULTS: Using an IRFA HCC orthotopic mouse model, we detected a higher level of m6 A reader YTH N6-methyladenosine RNA binding protein 1-3 (YTHDF1) in the sublethal-heat-exposed transitional zone close to the ablation center than that in the farther area. In addition, we validated the increased m6 A modification and elevated YTHDF1 protein level in sublethal-heat-treated HCC cell lines, HCC patient-derived xenograft (PDX) mouse model, and patients' HCC tissues. Functionally, gain-of-function/loss-of-function assays showed that YTHDF1 promotes HCC cell viability and metastasis. Knockdown of YTHDF1 drastically restrains the tumor metastasis evoked by sublethal heat treatment in tail vein injection lung metastasis and orthotopic HCC mouse models. Mechanistically, we found that sublethal heat treatment increases epidermal factor growth receptor (EGFR) m6 A modification in the vicinity of the 5' untranslated region and promotes its binding with YTHDF1, which enhances the translation of EGFR mRNA. The sublethal-heat-induced up-regulation of EGFR level was further confirmed in the IRFA HCC PDX mouse model and patients' tissues. Combination of YTHDF1 silencing and EGFR inhibition suppressed the malignancies of HCC cells synergically. CONCLUSIONS: The m6 A-YTHDF1-EGFR axis promotes HCC progression after IRFA, supporting the rationale for targeting m6 A machinery combined with EGFR inhibitors to suppress HCC metastasis after RFA.


Asunto(s)
Carcinoma Hepatocelular/cirugía , Neoplasias Hepáticas/cirugía , Procesamiento Postranscripcional del ARN/efectos de la radiación , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/efectos de la radiación , Ablación por Radiofrecuencia/efectos adversos , Animales , Carcinoma Hepatocelular/genética , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Receptores ErbB/genética , Receptores ErbB/metabolismo , Receptores ErbB/efectos de la radiación , Regulación Neoplásica de la Expresión Génica , Respuesta al Choque Térmico/efectos de la radiación , Humanos , Neoplasias Hepáticas/genética , Metilación/efectos de la radiación , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Procesamiento Postranscripcional del ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Insuficiencia del Tratamiento
19.
Cancer Lett ; 503: 1-10, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33444692

RESUMEN

Ablative treatment evokes antitumor immunity, but knowledge on the emerging irreversible electroporation (IRE)-induced immunity in hepatocellular carcinoma (HCC) is limited. To investigate the immune effects induced by IRE and its role in preventing post-ablation HCC progression, a C57BL/6J mouse model bearing subcutaneous H22 hepatoma was employed. IRE treatment significantly suppresses HCC growth, and treated mice are tumor-free after secondary tumor injection and show increased splenic interferon-gamma (IFN-γ)+CD8+ T cells. Additionally, more CD8+ T and dendritic cells, but not CD4+ T, B or NK cells, infiltrate into peri-ablation zones after IRE at day 7. Depletion of CD8+ T cells induces local tumor regrowth and distant metastasis after IRE. Vaccination using IRE-processed H22 lysates prevents tumorigenesis in mice, suggesting a protective immune response. IRE also alleviates immunosuppression by reducing local and splenic Treg and PD-1+ T cells. Regarding mechanism, IRE induces cell necrosis and significant release of danger-associated molecular patterns including ATP, high mobility group box 1 and calreticulin that are pivotal to CD8+ T cell immunity. Together, IRE is a promising approach to evoke CD8+ T cell immunity, which help prevent post-ablation HCC progression.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/terapia , Interferón gamma/metabolismo , Neoplasias Hepáticas/terapia , Ablación por Radiofrecuencia/métodos , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Dendríticas/inmunología , Progresión de la Enfermedad , Electroporación , Células Hep G2 , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
ACS Omega ; 5(14): 8272-8282, 2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32309738

RESUMEN

Binder, as one of the key components, plays a crucial role in improving the capacity and cycling performance of lithium-sulfur (Li-S) batteries. In this work, commercially available, low-cost, water-soluble polyvinyl alcohol (PVA) has been systematically investigated as a functional polymer binder for high-sulfur-loading cathodes, with the aim of enhancing sulfur utilization, reducing capacity decay, and extending cycling life of the cathodes. In comparison with polyvinylidene fluoride as a conventional binder, PVA shows a valuable polysulfide entrapping ability and a much stronger binding strength. Its superior polysulfide entrapping ability has been verified through theoretical density functional theory calculations and an experimental ex situ adsorption study. In electrochemical Li-S battery performance evaluation, at a sulfur loading density of 3.5 mg cm-2, the sulfur cathode assembled with the PVA binder displays at 0.5 C a very slow capacity decay of only 0.010% per cycle over 250 cycles. Additionally, the strong binding strength of PVA allows the fabrication of thick sulfur cathodes with a high sulfur loading density of 10.5 mg cm-2, which shows a high areal capacity of 4.0 mA h cm-2 and a high cycling stability (capacity decay of 0.1% per cycle). In consideration of the superior capacity retention and cycling performance of its enabled cathodes, the cost-effective PVA is a promising candidate for high-sulfur-loading cathodes in practical applications.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA