Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Anal Chem ; 95(18): 7271-7277, 2023 May 09.
Article in English | MEDLINE | ID: mdl-37103938

ABSTRACT

Label-free imaging of nanoscale targets with intrinsic properties is crucial for chemistry, physics, and life science to unveil the underlying mechanisms. Plasmonic imaging techniques are particularly attractive because they allow real-time imaging, providing insights into nanoscale detection and nanocatalysis. Here, we present a high-resolution plasmonic imaging method that is capable of imaging nanomaterials with high morphological fidelity and high throughput. We demonstrate that this approach allows for high-resolution plasmonic imaging of various nanomaterials ranging from nanoparticles and nanowires to two-dimensional nanomaterials and accurate tracking of the interfacial dynamics of nanoparticles. Given the experimental simplicity and capacity for label-free and real-time imaging of nanomaterials with high spatial resolution and high throughput, this approach can serve as a promising platform for characterizing nanomaterials at the single-particle level.

2.
BMC Cancer ; 23(1): 275, 2023 Mar 27.
Article in English | MEDLINE | ID: mdl-36973678

ABSTRACT

BACKGROUND: Inhibition of Serum Amyloid A-like 1 (SAAL1) expression could inhibit cancer progression and improve the prognosis of cancer patients. At present, the correlation between SAAL1 and lung adenocarcinoma (LAC) remains unclear. Therefore, this study surveyed the worth and pathway of SAAL1 in LAC progression and immunity. METHODS: Bioinformatics and immunohistochemistry were used to identify the SAAL1 expression in LAC. The roles of SAAL1 expression in the existence values of LAC patients were explored, and the nomograms were constructed. Clinical values of SAAL1 co-expressed genes were evaluated by COX regression, survival, and Receiver operating characteristic (ROC) analysis. EDU and western blotting methods were used to inquiry the functions and pathways of the SAAL1 in cell growths. The correlation between the SAAL1 level and immune microenvironment was visualized using correlation research. RESULTS: SAAL1 level was elevated in LAC tissues, and was observed in cancer tissues of dead patients. SAAL1 overexpression had something to do with shorter overall survival, progression-free interval, and disease-specific survival in LAC. The area under the curve of SAAL1 was 0.902 in normal tissues and cancer tissues. Inhibition of SAAL1 expression could inhibit cancer cell proliferation, which may be related to the decreased expression of cyclin D1 and Bcl-2 proteins. In LAC, SAAL1 level had something to do with stromal, immune, and estimate scores, and correlated with macrophages, T cells, Th2 cells, CD8 T cells, NK CD56dim cells, DC, eosinophils, NK CD56bright cells, pDC, iDC, cytotoxic cells, Tgd, aDC cells, B cells, Tcm, and TFH levels. SAAL1 overexpression had something to do with existence values and the immunity in LAC. CONCLUSIONS: Inhibition of SAAL1 expression could regulate cancer growth via cyclin D1 and Bcl-2. SAAL1 is a promising prognostic biomarker in LAC patients.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Cyclin D1 , Prognosis , Lung Neoplasms/pathology , Adenocarcinoma of Lung/genetics , Biomarkers, Tumor/genetics , Tumor Microenvironment
3.
Front Immunol ; 13: 986447, 2022.
Article in English | MEDLINE | ID: mdl-36544782

ABSTRACT

Immunotherapy can improve the survival of patients with advanced lung squamous cell carcinoma (LUSC). T cytotoxic cells are one of the main members of the immune microenvironment. Herein, we aimed to identify the roles of T-cell cytotoxic markers interleukin 18 (IL18) receptor 1 (IL18R1) in the LUSC progression using bioinformatics, clinical tissue specimen, and cell experiment. We assessed the association between the IL18R1 expression and immune infiltration and IL18R1-related competing RNA network. The IL18R1 expression was downregulated in the LUSC tissues. The IL18R1 expression downregulation was associated with diagnosis and short overall survival and disease-specific survival, and it was also an independent risk factor for dismal survival time in LUSC. IL18R1-related nomograms predicted the survival time of patients with LUSC. IL18R1 overexpression inhibited the proliferation, migration, and invasion of LUSC cells. The IL18R1 expression was significantly associated with the microenvironment (stromal, immune, and estimate scores), immune cells (such as the T cells, cytotoxic cells, CD8 T cells), and immune cell markers (such as the CD8A, PD-1, and CTLA4) in LUSC. AC091563.1 and RBPMS-AS1 downregulation was positively associated with the IL18R1 expression, negatively associated with the miR-128-3p expression, and associated with short disease-specific survival and progression in LUSC. In conclusion, IL18R1 was significantly downregulated and associated with the prognosis and immune microenvironment. IL18R1 overexpression inhibits the growth and migration of cancer cells in LUSC. Furthermore, AC091563.1 and RBPMS-AS1 might compete with IL18R1 to bind miR-128-3p for participating in LUSC progression. These results showed that IL18R1 is a biomarker for evaluating the prognosis of patients with LUSC.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , MicroRNAs , Humans , Down-Regulation , Prognosis , CD3 Complex , Interleukin-18 Receptor alpha Subunit , Lung Neoplasms/genetics , Cell Proliferation , Lung , MicroRNAs/genetics , Tumor Microenvironment
4.
Front Oncol ; 12: 960866, 2022.
Article in English | MEDLINE | ID: mdl-36276113

ABSTRACT

The roles and mechanisms of T-cell receptor (TCR)-associated transmembrane adaptor 1 (TRAT1) in lung adenocarcinoma (LAC) have not yet been reported in the relevant literature. Therefore, this study aimed to understand the roles and mechanisms of TRAT1 in LAC using bioinformatics and in vitro experiments. TRAT1 expression levels in LAC samples were analysed using various databases. TRAT1 co-expressed genes were acquired by the correlation analysis of LAC tissues. The functional mechanisms and protein network of TRAT1 co-expressed genes were analysed using bioinformatics analysis. The expression of TRAT1 was activated in LAC cells, and the roles of TRAT1 overexpression in the growth and migration of cancer cells was investigated using flow cytometry, Cell Counting Kit-8 (CCK-8), and migration and invasion assays. The relationship between TRAT1 overexpression, the immune microenvironment, and RNA modification was evaluated using correlation analysis. TRAT1 expression levels were significantly abnormal at multiple mutation sites and were related to the prognosis of LAC. TRAT1 co-expressed genes were involved in cell proliferation, adhesion, and differentiation, and TRAT1 overexpression significantly inhibited cell viability, migration, and invasion and promoted apoptosis of A549 and H1299 cells, which might be related to the TCR, B cell receptor (BCR), MAPK, and other pathways. TRAT1 expression levels were significantly correlated with the ESTIMATE, immune, and stromal scores in the LAC microenvironment. Additionally, TRAT1 expression levels were significantly correlated with the populations of B cells, CD8 T cells, cytotoxic cells, and other immune cells. TRAT1 overexpression was significantly correlated with the expression of immune cell markers (such as PDCD1, CD2, CD3E) and genes involved in RNA modification (such as ALKBH1, ALKBH3, ALKBH5). In conclusions, TRAT1 overexpression inhibited the growth and migration of LAC cells, thereby delaying cancer progression, and was correlated with the LAC microenvironment and RNA modifications.

5.
J Org Chem ; 82(11): 5652-5662, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28504515

ABSTRACT

Under the effect of a catalytic amount of Au(I) complex, 4-O-(2-cyclopropylethynyl)benzoyl-(epi)-podophyllotoxins, easily prepared via dehydrative condensation between (epi)-podophyllotoxin and ortho-cyclopropylethynylbenzoic acid, could efficiently couple with a variety of nucleophiles including alcohol, phenol, aniline, and carbon nucleophiles, all to provide (epi)-podophyllotoxin derivatives. Thus, the first catalytic and lignan-activation-based approach for (epi)-podophyllotoxin derivatization was established. Based on the new methodology, as well as the judicious choice of N3, AZMB, and Cbz protecting groups, an efficient approach forward was set. NK-611, an antitumoral agent at a phase II clinical trial was established, featuring an in situ anomerization of the hemiacetal OHs in the critical condensation step. Commencing from easily available starting material, the target molecule was obtained using the longest linear sequence of six steps and a 38% overall yield.

SELECTION OF CITATIONS
SEARCH DETAIL
...