Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Nanoscale ; 16(16): 8119-8131, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38567547

ABSTRACT

Electrocatalytic CO2 reduction (CO2RR) has emerged as a promising approach for converting CO2 into valuable chemicals and fuels to achieve a sustainable carbon cycle. However, the development of efficient electrocatalysts with high current densities and superior product selectivity remains a significant challenge. In this study, we present the synthesis of a porous nitrogen-doped carbon nanosheet loaded with heterostructured Ni/Ni3ZnC0.7 nanoparticles through a facile hydrothermal-calcination method (Ni/Ni3ZnC0.7-NC). Remarkably, the Ni/Ni3ZnC0.7-NC catalyst exhibits outstanding performance towards CO2RR in an H-cell, demonstrating a high CO faradaic efficiency of 92.47% and a current density (jCO) of 15.77 mA cm-2 at 0.87 V vs. RHE. To further explore its potential industrial applications, we constructed a flow cell and a rechargeable Zn-CO2 flow cell utilizing the Ni/Ni3ZnC0.7-NC catalyst as the cathode. Impressively, not only does the Ni/Ni3ZnC0.7-NC catalyst achieve an industrial high current density of 254 mA cm-2 at a voltage of -1.19 V vs. RHE in the flow cell, but it also exhibits a maximum power density of 4.2 mW cm-2 at 22 mA cm-2 in the Zn-CO2 flow cell, while maintaining excellent rechargeability. Density functional theory (DFT) calculations indicate that Ni/Ni3ZnC0.7-NC possesses more spontaneous reaction pathways for CO2 reduction to CO, owing to its heterogeneous structure in contrast to Ni3ZnC0.7-NC and Ni-NC. Consequently, Ni/Ni3ZnC0.7-NC demonstrates accelerated CO2RR reaction kinetics, resulting in improved catalytic activity and selectivity for CO2RR.

2.
J Immunother Cancer ; 11(7)2023 07.
Article in English | MEDLINE | ID: mdl-37524506

ABSTRACT

The treatment of B cell malignancies has dramatically changed with the introduction of immunotherapy, especially chimeric antigen receptor T (CAR-T) cell therapy. However, only limited efficacy is observed in acute myeloid leukaemia (AML). In the study, We detected CD123 and CLL-1 expression on leukaemia cells from Relapsed/Refractory AML (R/R AML) patients. Then, we constructed anti-CD123 CAR and CLL-1 CAR with different co-stimulation domains (CD28 or 4-1BB) and detected their anti-AML effects. To increase the efficacy of CAR-T cell therapy, we tested different strategies, including application of combined checkpoint inhibitors and histone deacetylase inhibitors (HDACi) in vivo and in vitro We found CD123 and CLL-1 were highly expressed on AML cells. The proportions of T cell subsets and NK cells involved in anti-tumour or anti-inflammation processes in AML patients significantly decreased when compared with healthy donors. Both CD123 CAR and CLL-1 CAR displayed specific anti-AML effects in vitro To improve the lysis effects of CAR-T cells, we combined CAR-T cell therapy with different agents. PD-1/PD-L1 antibodies only slightly improved the potency of CAR-T cell therapy (CD123 CAR-T 60.92% ± 2.9087% vs. 65.43% ± 2.1893%, 60.92% ± 2.9087% vs. 67.43% ± 3.4973%; 37.37% ± 3.908% vs. 41.89% ± 5.1568%, 37.37% ± 3.908% vs. 42.84% ± 4.2635%). However, one HDACi (valproic acid [VPA]) significantly improved CAR-T cell potency against AML cells (CLL-1 CAR-T 34.97% ± 0.3051% vs. 88.167% ± 1.5327%, p < 0.0001; CD123 CAR-T 26.87% ± 2.7010% vs. 82.56% ± 3.086%, p < 0.0001 in MV411; CLL-1 CAR-T 78.77% ± 1.2061% vs. 93.743% ± 1.2333%, p < 0.0001; CD123 CAR-T 64.10% ± 1.5130% vs. 94.427% ± 0.142%, p = 0.0001 in THP-1). Combination therapy prolonged the overall survival of mice when compared with single CD123 CAR-T cell therapy (median survival: 180 days vs. unfollowed). A possible mechanism is that activated CD8+T cells upregulate natural-killer group 2 member D (NKG2D), and VPA upregulates NKG2D ligand expression in AML cells, contributing to NKG2D-mediated cytotoxicity of CAR-T cells against tumour cells. In conclusion, CD123 and CLL-1 are promising targets for AML CAR-T cell therapy. A combination of VPA pre-treatment and CAR-T against AML exhibits synergic effects.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Animals , Mice , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Receptors, Chimeric Antigen/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Cell Line, Tumor , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , T-Lymphocytes
SELECTION OF CITATIONS
SEARCH DETAIL