Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 63(16): e202400952, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38372593

ABSTRACT

Cu-based catalysts have been identified as the most promising candidates for generation of C2+ products in electrochemical CO2 reduction reaction. Defect engineering in catalysts is a widely employed strategy for promoting C-C coupling on Cu. However, comprehensive understanding of defect structure-to-activity relationship has not been obtained. In this study, controllable defects generation is achieved, which leads to a series of Cu-based catalysts with various phase mixing degrees. It is observed that the Faradaic efficiency toward C2+ products increases with the phase mixing degree, reaching 81 % at maximum. In situ infrared absorption spectroscopy reveals that the catalysts with higher phase mixing degree tend to form *CO more easily and possess higher retention of *CO under high overpotential window, thereby promoting C-C coupling. This work sheds new light on the relationship between defects and C-C coupling, and the rational developed of more advanced Cu-base catalysts.

2.
Angew Chem Int Ed Engl ; 63(9): e202313858, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38185801

ABSTRACT

Catalysts involving post-transition metals have shown almost invincible performance on generating formate in electrochemical CO2 reduction reaction (CO2 RR). Conversely, Cu without post-transition metals has struggled to achieve comparable activity. In this study, a sulfur (S)-doped-copper (Cu)-based catalyst is developed, exhibiting excellent performance in formate generation with a maximum Faradaic efficiency of 92 % and a partial current density of 321 mA cm-2 . Ex situ structural elucidations reveal the presence of abundant grain boundaries and high retention of S-S bonds from the covellite phase during CO2 RR. Furthermore, thermodynamic calculations demonstrate that S-S bonds can moderate the binding energies with various intermediates, further improving the activity of the formate pathway. This work is significant in modifying a low-cost catalyst (Cu) with a non-metallic element (S) to achieve comparable performance to mainstream catalysts for formate generation in industrial grade.

3.
Sci Transl Med ; 15(678): eabl7895, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36630483

ABSTRACT

Pancreatic and lung cancers frequently develop resistance to chemotherapy-induced cell apoptosis during the treatment, indicating that targeting nonapoptotic-related pathways, such as pyroptosis, can be an alternative cancer treatment strategy. Pyroptosis is a gasdermin-driven lytic programmed cell death triggered by inflammatory caspases when initiated by canonical or noncanonical pathways that has been recently seen as a potential therapeutic target in cancer treatment. However, overcoming chemoresistance in cancers by modulating pyroptosis has not been explored. Here, we demonstrate that ß5-integrin represses chemotherapy-induced canonical pyroptosis to confer cancer chemoresistance through ASAH2-driven sphingolipid metabolic reprogramming. Clinically, high ß5-integrin expression associates with poor patient prognosis and chemotherapeutic responses in cancers. In addition, chemoresistant cells in vitro fail to undergo chemotherapy-induced pyroptosis, which is controlled by ß5-integrin. Mechanistically, proteomic and lipidomic analyses indicate that ß5-integrin up-regulates sphingolipid metabolic enzyme ceramidase (ASAH2) expression through Src-signal transducer and activator of transcription 3 (STAT3) signaling, which then reduces the metabolite ceramide concentration and subsequent ROS production to prohibit chemotherapy-induced canonical pyroptosis. Using cancer cell lines, patient-derived tumor organoids, and orthotopic lung and pancreatic animal models, we show that administration of a Src or ceramidase inhibitor rescues the response of chemoresistant pancreatic and lung cancer cells to chemotherapy by reactivating pyroptosis in vitro and in vivo. Overall, our results suggest that pyroptosis-based therapy is a means to improve cancer treatment and warrants further investigation.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Pancreatic Neoplasms , Proto-Oncogene Proteins pp60(c-src) , Pyroptosis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Integrins/metabolism , Lung/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proteomics , Pyroptosis/drug effects , Proto-Oncogene Proteins pp60(c-src)/drug effects , Proto-Oncogene Proteins pp60(c-src)/metabolism , Humans , Integrin beta Chains/metabolism , STAT3 Transcription Factor/metabolism , Ceramidases/metabolism , Pancreatic Neoplasms
4.
Pharmacol Res ; 187: 106558, 2023 01.
Article in English | MEDLINE | ID: mdl-36410675

ABSTRACT

Dysregulated sphingolipid metabolism contributes to ER+ breast cancer progression and therapeutic response, whereas its underlying mechanism and contribution to tamoxifen resistance (TAMR) is unknown. Here, we establish sphingolipid metabolic enzyme CERK as a regulator of TAMR in breast cancer. Multi-omics analysis reveals an elevated CERK driven sphingolipid metabolic reprogramming in TAMR cells, while high CERK expression associates with worse patient prognosis in ER+ breast cancer. CERK overexpression confers tamoxifen resistance and promotes tumorigenicity in ER+ breast cancer cells. Knocking out CERK inhibits the orthotopic breast tumor growth of TAMR cells while rescuing their tamoxifen sensitivity. Mechanistically, the elevated EHF expression transcriptionally up-regulates CERK expression to prohibit tamoxifen-induced sphingolipid ceramide accumulation, which then inhibits tamoxifen-mediated repression on PI3K/AKT dependent cell proliferation and its driven p53/caspase-3 mediated apoptosis in TAMR cells. This work provides insight into the regulation of sphingolipid metabolism in tamoxifen resistance and identifies a potential therapeutic target for this disease.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Tamoxifen , Female , Humans , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Estrogen/metabolism , Sphingolipids , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
5.
J Am Chem Soc ; 144(44): 20372-20384, 2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36283038

ABSTRACT

The poor durability of Pt-based nanoparticles dispersed on carbon black is the challenge for the application of long-life polymer electrolyte fuel cells. Recent work suggests that Fe- and N-codoped carbon (Fe-N-C) might be a better support than conventional high-surface-area carbon. In this work, we find that the electrochemical surface area retention of Pt/Fe-N-C is much better than that of commercial Pt/C during potential cycling in both acidic and basic media. In situ inductively coupled plasma mass spectrometry studies indicate that the Pt dissolution rate of Pt/Fe-N-C is 3 times smaller than that of Pt/C during cycling. Density functional theory calculations further illustrate that the Fe-N-C substrate can provide strong and stable support to the Pt nanoparticles and alleviate the oxide formation by adjusting the electronic structure. The strong metal-substrate interaction, together with a lower metal dissolution rate and highly stable support, may be the reason for the significantly enhanced stability of Pt/Fe-N-C. This finding highlights the importance of carbon support selection to achieve a more durable Pt-based electrocatalyst for fuel cells.

6.
Environ Sci Pollut Res Int ; 29(51): 76728-76738, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35670935

ABSTRACT

The adsorption behaviors of cellulose and lignin biochar depend on the evolution of their oxygen-containing groups to some extent. In this study, cellulose-rich pakchoi and lignin-rich corncob were selected to prepare the pyrolytic biochar at variable temperatures, named PBC and CBC, respectively. Their structure-function relationships were in-depth studied via the combination of the adsorption experiments of U(VI) and comprehensive spectral analyses. The maximal adsorption capacity of PBC 300, obtained at 300 °C, was measured as 46.62 mg g-1 for U(VI), which was ⁓1.3 times higher than 35.60 mg g-1 of CBC 300. U(VI) adsorption on PBC and CBC were predominantly ascribed to the coordination interaction between oxygen-containing groups and U(VI). Interestingly, the main complexation groups were distinct in both biochars due to the different inherent evolutions of cellulose and lignin. Volatile d-glucose chains in cellulose were apt to degrade rapidly, and the formed carboxyls acted as the most important sites in PBC. However, the stable aromatic network in lignin led to a slow degradation, and more hydroxyls thus remained in CBC, which controlled U(VI) adsorption. In this study, we obtained greatly cost-effective adsorbents of U(VI) and provided some essential insights into understanding the structural evolution-function relationship of cellulose and lignin biochar.


Subject(s)
Cellulose , Lignin , Adsorption , Lignin/chemistry , Oxygen , Charcoal/chemistry , Glucose
7.
Nat Commun ; 12(1): 6011, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650057

ABSTRACT

Defective pericyte-endothelial cell interaction in tumors leads to a chaotic, poorly organized and dysfunctional vasculature. However, the underlying mechanism behind this is poorly studied. Herein, we develop a method that combines magnetic beads and flow cytometry cell sorting to isolate pericytes from tumors and normal adjacent tissues from patients with non-small cell lung cancer (NSCLC) and hepatocellular carcinoma (HCC). Pericytes from tumors show defective blood vessel supporting functions when comparing to those obtained from normal tissues. Mechanistically, combined proteomics and metabolic flux analysis reveals elevated hexokinase 2(HK2)-driven glycolysis in tumor pericytes, which up-regulates their ROCK2-MLC2 mediated contractility leading to impaired blood vessel supporting function. Clinically, high percentage of HK2 positive pericytes in blood vessels correlates with poor patient overall survival in NSCLC and HCC. Administration of a HK2 inhibitor induces pericyte-MLC2 driven tumor vasculature remodeling leading to enhanced drug delivery and efficacy against tumor growth. Overall, these data suggest that glycolysis in tumor pericytes regulates their blood vessel supporting role.


Subject(s)
Blood Vessels/abnormalities , Glycolysis , Hexokinase/metabolism , Neoplasms, Vascular Tissue/metabolism , Pericytes/metabolism , A549 Cells , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Hexokinase/genetics , Humans , Mice , Mice, Inbred C57BL , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Neoplasms/metabolism , Neoplasms, Vascular Tissue/drug therapy , Neoplasms, Vascular Tissue/genetics , Neoplasms, Vascular Tissue/pathology , Tumor Microenvironment/physiology , Up-Regulation , rho-Associated Kinases
8.
Front Cell Dev Biol ; 9: 676342, 2021.
Article in English | MEDLINE | ID: mdl-34179005

ABSTRACT

Pericytes (PCs), known as mural cells, play an important blood vessel (BV) supporting role in regulating vascular stabilization, permeability and blood flow in microcirculation as well as blood brain barrier. In carcinogenesis, defective interaction between PCs and endothelial cells (ECs) contributes to the formation of leaky, chaotic and dysfunctional vasculature in tumors. However, recent works from other laboratories and our own demonstrate that the direct interaction between PCs and other stromal cells/cancer cells can modulate tumor microenvironment (TME) to favor cancer growth and progression, independent of its BV supporting role. Furthermore, accumulating evidence suggests that PCs have an immunomodulatory role. In the current review, we focus on recent advancement in understanding PC's regulatory role in the TME by communicating with ECs, immune cells, and tumor cells, and discuss how we can target PC's functions to re-model TME for an improved cancer treatment strategy.

9.
Adv Mater ; 33(23): e2100407, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33909930

ABSTRACT

Although oxygen vacancies (Ov s) play a critical role for many applications of metal oxides, a controllable synthetic strategy for anisotropic Ov s remains a great challenge. Here, a novel strategy is proposed to achieve the regional dual structure with anisotropic Ov s at both the surface and in the interior of TiO2 by constructing amorphous domains. The as-prepared black TiO2 with amorphous domains exhibits superior activity in degrading rhodamine B (RhB) solutions, which can instantly decompose RhB with just a shake. First-principle simulations reveal that subsurface Ov s in TiO2 are energetically favored, resulting in the formation of amorphous domains in the interior region via diffusion of surface-formed Ov s into the subsurface. The stable Ov -induced amorphous domains in TiO2 with enhanced catalytic performances provide a scalable strategy to practical Ov engineering in functional metal oxides.

10.
EBioMedicine ; 66: 103301, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33813137

ABSTRACT

BACKGROUND: Sphingolipid metabolism is among the top dysregulated pathways in non-small cell lung carcinomas (NSCLC). However, the molecular control of sphingolipid metabolic reprogramming in cancer progression remains unclear. METHODS: We first determined the correlation between sphingolipid metabolic gene expression and patient prognosis. We then carried out sphingolipidomics analysis of health individual and NSCLC patient sera as well as B3GNT5 and GAL3ST1 genetically perturbed NSCLC cell lines. We used these cell lines to perform tumorigenesis study to determine the cellular role of B3GNT5 and GAL3ST1 in cancer growth and progression. FINDINGS: The expression of B3GNT5 and GAL3ST1 among sphingolipid metabolic enzymes is most significantly associated with patient prognosis, whilst sphingolipidomics analysis of healthy individual and NSCLC patient sera identifies their metabolites, lacto/neolacto-series glycosphingolipid and sulfatide species, as potential biomarkers that were more effective than current clinical biomarkers for staging patients. Further network analysis of the sphingolipidomes reveals a circular network of coregulated sphingolipids, indicating that the lacto/neolacto-series glycosphingolipid/sulfatide balance functions as a checkpoint to determine sphingolipid metabolic reprograming during patient progression. Sphingolipidomics analysis of B3GNT5/GAL3ST1 genetically perturbed NSCLC cell lines confirms their key regulatory role in sphingolipid metabolism, while B3GNT5 and GAL3ST1 expression has an opposite role on tumorigenesis. INTERPRETATION: Our results provide new insights whereby B3GNT5 and GAL3ST1 differentially regulate sphingolipid metabolism in lung cancer growth and progression. FUNDING: This work was supported by the Natural Science Foundation of China (81872142, 81920108028); Guangzhou Science and Technology Program (201904020008); Guangdong Science and Technology Department (2020A0505100029, 2019A1515011802, 2020A1515011280, 2020B1212060018, 2020B1212030004); China Postdoctoral Science Foundation (2019M650226, 2019M650227).


Subject(s)
Lipid Metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Sphingolipids/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Computational Biology/methods , Databases, Genetic , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Lipidomics/methods , Lung Neoplasms/etiology , Lung Neoplasms/mortality , Mice , Neoplasm Staging , Prognosis
11.
Cancer Lett ; 500: 228-243, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33309857

ABSTRACT

Aberrant glycosylation in pancreatic cancer has been linked to cancer development, progression and chemoresistance. However, the role of glycogene, such as galactosyltransferase, in pancreatic cancer remains unknown. Herein, we establish beta-1.4-galactosyltransferase 1 (B4GALT1) as a clinical marker and regulator of chemoresistance. Clinically, high B4GALT1 expression correlates with poor survival, enhanced tumor size, increased lymph node metastasis, elevated cancer progression and enhanced incidence of relapse in PDAC patients. Expression of B4GALT1 is up-regulated in gemcitabine resistant patient derived organoids as well as chemoresistant cancer cell lines, while genetic perturbation of its expression in PDAC cell lines regulates cancer progression and chemoresistance. Mechanistically, we show that elevated p65 activity transcriptionally up-regulates B4GALT1 expression, which then interacts with and stabilizes cyclin dependent kinase 11 isomer CDK11p110 protein via N-linked glycosylation, in order to promote cancer progression and chemoresistance. Finally, depletion of B4GALT1 rescues the response of chemoresistant cells to gemcitabine in an orthotopic PDAC model. Overall, our data uncovers a mechanism by which p65-B4GALT1-CDK11p110 signalling axis determines cancer progression and chemoresistance, providing a new therapeutic target for an improved pancreatic cancer treatment.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Cyclin-Dependent Kinases/genetics , Galactosyltransferases/genetics , Transcription Factor RelA/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glycosylation/drug effects , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Organoids/drug effects , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...