Xiaosong Li’s research while affiliated with First Affiliated Hospital of China Medical University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (58)


Shows the intricate interaction among different populations of immune cells in the context of HBV-related HCC. The primary emphasis is tumor-associated macrophages (TAMs) and CD8+ T cells as well as their interactions with other immune cells, such as Neutrophils, Natural killer (NK) cells and Dendritic cells (DCs). The figure is created with BioRender.com.
Shows the development of macrophages in HBV-HCC, showing their diverse origins and the mechanism of M1/M2 polarization, as well as their role in cancer. The figure is created with BioRender.com.
Illustrates the dual nature of CD8+ T cells in the context of HBV-HCC. It is establishing a balance between viral management, anticancer activity, and exhaustion against cancer.
This figure highlights the interplay between TAMs and CD8+ T cells in the TME of HBV-related HCC. TAMs suppress CD8+ T cell function by several mechanisms such as direct and indirect macrophage events, arginine deprivation, and hypoxia.
Advances in checkpoint inhibitor in HBV-HCC.
Tumor-associated macrophages and CD8+ T cells: dual players in the pathogenesis of HBV-related HCC
  • Literature Review
  • Full-text available

October 2024

·

54 Reads

·

Binli Mao

·

Juan Hu

·

[...]

·

Xiaosong Li

HBV infection is a key risk factor for the development and progression of hepatocellular carcinoma (HCC), a highly invasive tumor, and is characterized by its persistent immunosuppressive microenvironment. This review provides an in-depth analysis of HBV-related HCC and explores the interactions between neutrophils, natural killer cells, and dendritic cells, examining their roles in regulating tumor-associated macrophages and CD8+ T cells and shaping the tumor microenvironment. Two critical players in the immunosuppressive milieu of HBV-related HCC are CD8+ T cells and tumor-associated macrophages (TAMs). The study explores how TAMs, initially recruited to combat infection, transform, adopting a tumor-promoting phenotype, turning against the body, promoting tumor cell proliferation, suppressing anti-tumor immunity, and assisting in the spread of cancer. Meanwhile, CD8+ T cells, crucial for controlling HBV infection, become dysfunctional and exhausted in response to persistent chronic viral inflammation. The review then dissects how TAMs manipulate this immune response, further depleting CD8+ T cell functions through mechanisms like arginine deprivation and creating hypoxic environments that lead to exhaustion. Finally, it explores the challenges and promising therapeutic avenues that target TAMs and CD8+ T cells, either separately or in combination with antiviral therapy and personalized medicine approaches, offering hope for improved outcomes in HBV-related HCC.

Download

Fig. 1. Main biding partners of the VISTA and the involved structures. VISTA interacts with VSIG3 at physiological pH, but acidic (pH~6.0) tumor microenvironment (TME) facilitates VISTA-expressing cells binding to PSGL-1 on T cell. Both interactions curtail T cell activities. Both associations reduce the activity of T cells [46]. Both engage with VISTA in the F68 and K70 of the C strand, the R86 and Q95 of the C-C′ loop, and the F-G loop (H154) [47].
Fig. 2. MDSCs create immunosuppressive TME and the TLR/MyD88 signaling on MDSC is downregulated by VISTA. MDSC primarily inhibits T cells by 1) increasing the expression of T cell immunoinhibitory receptors, 2) secreting immunosuppressive cytokines, 3) depriving T cells of critical amino acids for activity and proliferation, 4) gathering Tregs, and 5) enhancing the generation of free radical. The TLR/MyD88-mediated communication is downregulated by VISTA. After TLRs are activated, the MyD88/IRAK1/4 complex recruits and triggers TRAF6, which in turn stimulates the MAPK/AP-1, IKK/NF-B, and MAPK pathways.
Fig. 3. VISTA regulates MDSC-mediated inhibition through various mechanisms. By increasing the expression of VISTA on MDSC, hypoxia improves the delivery of co-inhibitory signals to T cells. VISTA inhibits TLR-mediated communication and the development of the MDSC C5a receptor on cell surfaces. The MDSC-mediated suppression enhancement through ARG1 also involves VISTA.
Targeting VISTA contributes to reducing MDSCs.
Eliminating a barrier: Aiming at VISTA, reversing MDSC-mediated T cell suppression in the tumor microenvironment

September 2024

·

29 Reads

·

3 Citations

Heliyon

Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by producing remarkable clinical outcomes for patients with various cancer types. However, only a subset of patients benefits from immunotherapeutic interventions due to the primary and acquired resistance to ICIs. Myeloid-derived suppressor cells (MDSCs) play a crucial role in creating an immunosuppressive tumor microenvironment (TME) and contribute to resistance to immunotherapy. V-domain Ig suppressor of T cell activation (VISTA), a negative immune checkpoint protein highly expressed on MDSCs, presents a promising target for overcoming resistance to current ICIs. This article provides an overview of the evidence supporting VISTA's role in regulating MDSCs in shaping the TME, thus offering insights into how to overcome immunotherapy resistance.




Characterization of Pt@ZIF‐8. a) Schematic illustration of synthetic process of Pt@ZIF‐8. b) TEM images of ZIF‐8. c–e) TEM images of Pt@ZIF‐8 nanoparticles with Pt contents of 0.59 mg (c, 400 nm),1.1 mg (d, 200 nm), and 1.47 mg (e, 100 nm), respectively. f) Hydrodynamic diameter of ZIF‐8 (97.4 ± 2.72 nm), 400 nm Pt@ZIF‐8 (417.7 ± 6.39 nm), 200 nm Pt@ZIF‐8 (224.3 ± 2.68 nm), and 100 nm Pt@ZIF‐8 (109.8 ± 0.53 nm). g) UV–vis absorption spectra of ZIF‐8 and Pt@ZIF‐8 with different sizes against TMB‐H2O2 solution. h) UV–vis absorption spectra of ZIF‐8 and Pt@ZIF‐8 synthesized under the same ZIF‐8 feedstock concentration. i) TEM images of 200 nm Pt@ZIF‐8 in water solution after 7 d. j) Hydrodynamic diameter of 200 nm Pt@ZIF‐8 in water solution after 7 d. k) Pore‐size distributions of ZIF‐8 calculated by the Horvath–Kawazoe method.
Characterization of Pt@ZIF‐8@PDA and Pt@ZIF‐8@PDA@Pt. a) Schematic illustration of synthetic process of Pt@ZIF‐8@PDA and Pt@ZIF‐8@PDA@Pt. b) UV–vis absorption spectra of the Pt@ZIF‐8@PDA synthesized with different mass proportions of PDA and Pt@ZIF‐8; inset: photographs of corresponding solution at a concentration of 2 mg mL⁻¹. c) UV–vis absorption spectra of the Pt@ZIF‐8@PDA, which synthesized with different mass ratios of PDA/Pt@ZIF‐8 against TMB‐H2O2 solution. d) Optical intensity of Pt@ZIF‐8@PDA with different mass ratios of PDA/Pt@ZIF‐8 before AEC catalyzed and AEC catalyzed enhancement on NC membrane; the concentration was 1 mg mL⁻¹. The error bar represents standard deviation (SD), n = 3. e) TEM images of Pt@ZIF‐8@PDA. f) TEM images of Pt@ZIF‐8@PDA@Pt. g) HAADF STEM and EDS mapping characterization of Pt@ZIF‐8@PDA@Pt. h) UV−vis absorption spectra of Pt@ZIF‐8, Pt@ZIF‐8@PDA, and Pt@ZIF‐8@PDA@Pt; inset: photograph of solutions at a concentration of 2 mg mL⁻¹. i) Hydrodynamic diameter of Pt@ZIF‐8 (228.3 ± 2.112 nm), Pt@ZIF‐8@PDA (245.1 ± 1.753 nm), and Pt@ZIF‐8@PDA@Pt (262.8 ± 3.279 nm). The error bar represents SD, n = 3. j) ζ potential of the Pt@ZIF‐8 (49.61 ± 0.7296 mV), Pt@ZIF‐8@PDA (−38.74 ± 1.314 mV), and Pt@ZIF‐8@PDA@Pt (43.9 ± 1.428 mV). The error bar represents SD, n = 3. k–m) XRD, XPS, and FT‐IR spectra of Pt@ZIF‐8, Pt@ZIF‐8@PDA, and Pt@ZIF‐8@PDA@Pt, respectively.
Catalytic properties and stability of Pt@ZIF‐8@PDA@Pt. a,b) UV–vis absorption spectra of Pt NPs, Pt@ZIF‐8, Pt@ZIF‐8@PDA, and Pt@ZIF‐8@PDA@Pt at the same Pt content against TMB concentration (0.2 m NaAc‐HAc buffer solution, pH 4.0, containing 1 × 10⁻³ m TMB) in a) the presence of 1 × 10⁻³ m H2O2 and b) absence of H2O2. c) Photographs and corresponding optical intensity of Pt@ZIF‐8@PDA@Pt before AEC catalyzed and AEC catalyzed enhancement on NC membrane; the concentration was 0.5 mg mL⁻¹. The error bar represents SD, n = 3. d) Photograp and e) corresponding absorbance values at 652 nm of TMB substrate solution catalyzed by Pt@ZIF‐8, Pt@ZIF‐8@PDA, and Pt@ZIF‐8@PDA@Pt of various concentrations at room temperature, respectively. The error bar represents SD, n = 3. f–h) Relative activity of Pt@ZIF‐8@PDA@Pt and free HRP after storage at 4 °C for a week f) and preservation in solution with g) different buffers and g) pH. The error bar represents SD, n = 3. i) Changes in relative activity of Pt@ZIF‐8@PDA@Pt at 45 °C for a week. The error bar represents SD, n = 3. j) Reproducibility of signal amplification by Pt@ZIF‐8@PDA@Pt assisted catalytic deposition via monitoring the changes in optical intensity before and after catalyzed AEC within 7 d. Pt@ZIF‐8@PDA@Pt at a concentration of 1 mg mL⁻¹ was deposited on the NC membrane as a T line. The error bar represents SD, n = 3. k) Change in hydrodynamic diameter and zeta potential of Pt@ZIF‐8@PDA@Pt within 21 d. The error bar represents SD, n = 3.
Analytical performance of the prepared Pt@ZIF‐8@PDA@Pt‐based LFIA test strips for NT‐proBNP detection. a) Photographs of test strips in response to different concentrations of NT‐proBNP before and after AEC catalytic amplification. b,c) Calibration curves of the T‐line intensity before and after AEC catalytic amplification, respectively. The error bar represents SD, n = 3. d) Selectivity of the Pt@ZIF‐8@PDA@Pt‐based LFIA test strips. The error bar represents SD, n = 3. e) Heat maps showing the results of NT‐proBNP detection in clinical serum samples using the Pt@ZIF‐8@PDA@Pt‐based LFIA. Values represented in the heat map are the concentrations of NT‐proBNP in each sample. f) Correlation analysis between the Pt@ZIF‐8@PDA@Pt‐based LFIA and the CLIA in quantifying the NT‐proBNP concentration, considering clinical serum samples.
Representation of the Pt@ZIF‐8@PDA@Pt‐immunolabeled lateral flow immunoassay for ultrasensitive detection of NT‐proBNP in serum samples.
Flexible Scaffold Modulation of Spatial Structure and Function of Hierarchically Porous Nanoparticle@ZIF‐8 Composites to Enhance Field Deployable Disease Diagnostics

Catalytic nanoparticle@metal‐organic framework (MOF) composites have attracted significant interest in point‐of‐care testing (POCT) owing to their prominent catalytic activity. However, the trade‐off between high loading efficiency and high catalytic activity remains challenging because high concentrations of nanoparticles tend to cause the misjoining and collapse of the MOFs. Herein, a facile strategy is reported to encapsulate high concentrations of platinum (Pt) nanoparticles into zeolitic imidazolate framework‐8 (ZIF‐8) using polydopamine (PDA) as a support for Pt@ZIF‐8 and as a flexible scaffold for further immobilization of Pt nanoparticles. The resulting composite (Pt@ZIF‐8@PDA@Pt) exhibits ultrahigh Pt nanoparticle loading efficiency, exceptional catalytic activity, stability, and a bright colorimetric signal. Following integration with lateral flow immunoassay (LFIA), the detection limits for pre‐ and post‐catalysis detection of B‐type natriuretic peptide (NT‐proBNP) are 0.18 and 0.015 ng mL⁻¹, respectively, representing a 6‐fold and 70‐fold improvement compared to gold nanoparticle‐based LFIA. Moreover, Pt@ZIF‐8@PDA@Pt‐based LFIA achieves 100% diagnostic sensitivity for NT‐proBNP in a cohort of 184 clinical samples.



Paclitaxel-induced Immune Dysfunction and Activation of Transcription Factor AP-1 Facilitate Hepatitis B Virus Replication

March 2024

·

17 Reads

·

1 Citation

Background and Aims Hepatitis B virus (HBV) reactivation is commonly observed in individuals with chronic HBV infection undergoing antineoplastic drug therapy. Paclitaxel (PTX) treatment has been identified as a potential trigger for HBV reactivation. This study aimed to uncover the mechanisms of PTX-induced HBV reactivation in vitro and in vivo, which may inform new strategies for HBV antiviral treatment. Methods The impact of PTX on HBV replication was assessed through various methods including enzyme-linked immunosorbent assay, dual-luciferase reporter assay, quantitative real-time PCR, chromatin immunoprecipitation, and immunohistochemical staining. Transcriptome sequencing and 16S rRNA sequencing were employed to assess alterations in the transcriptome and microbial diversity in PTX-treated HBV transgenic mice. Results PTX enhanced the levels of HBV 3.5-kb mRNA, HBV DNA, HBeAg, and HBsAg both in vitro and in vivo. PTX also promoted the activity of the HBV core promoter and transcription factor AP-1. Inhibition of AP-1 gene expression markedly suppressed PTX-induced HBV reactivation. Transcriptome sequencing revealed that PTX activated the immune-related signaling networks such as IL-17, NF-κB, and MAPK signaling pathways, with the pivotal common key molecule being AP-1. The 16S rRNA sequencing revealed that PTX induced dysbiosis of gut microbiota. Conclusions PTX-induced HBV reactivation was likely a synergistic outcome of immune suppression and direct stimulation of HBV replication through the enhancement of HBV core promoter activity mediated by the transcription factor AP-1. These findings propose a novel molecular mechanism, underscoring the critical role of AP-1 in PTX-induced HBV reactivation.


Oxytocin alleviates liver fibrosis via hepatic macrophages

February 2024

·

56 Reads

·

7 Citations

JHEP Reports

Background & Aims Previous studies demonstrated oxytocin treatment effectiveness in reducing mortality and reversing liver fibrosis in mice. However, the underlying mechanism remains obscure given the absence of oxytocin receptor expression in hepatic stellate cells, the primary liver fibrosis effector cells. Methods A comprehensive map of cell populations in fibrotic liver was generated using single-cell sequencing. The map enabled our study of the target cells of oxytocin action in the liver in more dimensions. Furthermore, we elucidated the oxytocin signaling system mechanism in hepatic macrophages using oxytocin receptor-specific knockout mice and liver fibrosis animal models. Results The carbon tetrachloride (CCl4)-induced hepatic fibrosis and bile duct ligation hepatic fibrosis mouse models demonstrated that oxytocin reversed hepatic fibrosis in mice. The mapped liver cell populations demonstrated that oxytocin promoted the phenotypic switch from Ly6high to Ly6Clow in myeloid-derived macrophages. The phenotypic control of oxytocin signaling system activation on the above phenotypic switch was validated using myeloid-specific OXTR knockout mice. Subsequent studies demonstrated that the calcium inward flow induced by oxytocin receptor activation activated the key orphan nuclear receptor NR4A1, which controls macrophage phenotypic switching. Specifically, calcium ions activated CREB, a key target regulator of NR4A1 expression. Conclusion The findings established hepatic macrophages as a hub responsible for the oxytocin-mediated alleviation of liver fibrosis. This study revealed a novel pathway where oxytocin regulates macrophage phenotype.



Figure 1. Illustration of cSDR ( A ) and CtSDR ( B ) . ( C ) The fluorescence curves for cSDR ( black line ) and CtSDR ( red line ) . 0.8 U / μl RNase inhibitor, 100 nM DS-12, Cas12a and crRNA, and 1 × NEBuffer 3.0, recorded by FS5 fluorescence spectrophotometer. ( D ) Typical SPR sensorgrams of CtSDR with different Cas12a / crRNA complex concentrations ( 10 0 0, 80 0, 50 0, 20 0 nM ) . 1 μM DS-12 ( SH ) , and 1 × NEBuffer 3.0. ( E ) Typical SPR sensorgrams of cSDR with different crRNA concentrations ( 10 0 0, 80 0, 50 0, 20 0 nM ) : 'a' shows the timepoint of react ant injection, and 'b' shows the timepoint of washing. 1 μM DS-12 ( SH ) , and 1 × NEBuffer 3.0. The corresponding K D values were evaluated with TraceDrawer. ( F ) Illustration of cSDR, CtSDR, and CtSDR-aCas14a of the Cas9 system ( first group ) , Cas12a system ( second group ) , and corresponding normalized FI of Cy3 ( G ) , respectively. Error bars represent standard deviations of three independent experiments **** t < 0.0 0 01, ** t < 0.01. 0.8 U / μl RNase inhibitor, 200 nM DS-21 / DS-Cas9, 200 nM Cas1 2a / Cas9 / Cas1 4a, 200 nM crRNA-1 4 / Inv-cr-1 2 / Inv-cr-9, 200 nM A-14 and 1 × NEBuffer 3.0, recorded by FS5 fluorescence spectrophotometer.
Figure 2. ( A ) Illustration of cSDR and CtSDR with different displacement directions and toehold base numbers. ( B ) The FI of Cy3 produced by cSDR and CtSDR with different toehold lengths ( range from 2 to 8 ) and locations ( 3 -toe and 5 -toe ) , and ( C ) the FI of FAM produced by the trans -cleavage of CtSDR with different toehold lengths ( range from 2 to 8 ) and locations ( 3 -toe and 5 -toe ) . 500 nM RP, 0.8 U / μl RNase inhibitor, 20 nM DS-3 -toe-n and DS-5 -toe-n, Cas12a and crRNA, and 1 × NEBuffer 3.0, recorded by FS5 fluorescence spectrophotometer. ( D ) The real-time fluorescence curve of Cy3 produced by cSDR and CtSDR with different toehold base numbers. 1 μM RP, 0.8 U / μl RNase inhibitor , 1 00 nM DS-3 -toe-n and DS-5 -toe-n, Cas12a and crRNA, and 1 × NEBuffer 3.0, recorded by 7500 real-time PCR. ( E ) The real-time fluorescence curve of FAM produced by the trans -cleavage of CtSDR with different toehold base numbers. Error bars represent the standard deviation of three independent experiments. 1 μM RP, 0.8 U / μl RNase inhibitor,
Figure 4. ( A ) Confocal microscopic images of MCF-7 cells ( a to c ) and MCF-10A cells ( d to f ) after incubation with target-initiated CtSDR for simultaneously detecting miR-21 and miR-155. The first column is Cy3 fluorescence with excitation at 556 nm, the second column is Cy5 fluorescence with e x citation at 643 nm, the third column is FAM fluorescence with e x citation at 492 nm, the f ourth column is DAPI fluorescence, and the last column is the merged fluorescence image. Scale bar = 25 μm. 2 μM Cas12a, 1 μM crRNA-21 / 155, 1 μM DS-21 / 155, 10 μM RP, 0.8 U / μl RNase inhibitor and 1 × NEBuffer 3.0. ( B ) Flo w-cytometry analy sis of CtSDR in MCF 7 and MCF 10A. 2 μM Cas12a, 1 μM crRNA-21 / 155, 1 μM DS-21 / 155, 10 μM RP, 0.4 U / μl RNase inhibitor and 1 × NEBuffer 3.0. ( C ) Confocal microscopic images of target-initiated CtSDR in MCF-7 cells after treatment with inhibitors and the inhibitor negative control ( inhibitor-NC ) . The first column is Cy3 fluorescence with excitation at 556 nm, the second column is Cy5 fluorescence with e x citation at 643 nm, the third column is FAM fluorescence with e x citation at 492 nm, the fourth column is DAPI fluorescence, and the last column is the merged fluorescence image. Scale bar = 25 μm. 2 μM Cas12a, 1 μM crRNA-21 / 155, 1 μM DS-21 / 155, 10 μM RP, 0.8 U / μl RNase inhibitor and
Figure 5. ( A ) Workflow comparison between the CtSDR-based imaging strategy and traditional ISH. ( B ) Fluorescence microscopic images of tissues after treatment with target-initiated CtSDR. The first row shows FAM fluorescence with excitation at 492 nm, the second row shows DAPI fluorescence, and the last row shows a merged fluorescence image. Scale bar = 80 μm. 2 μM Cas12a, 1 μM crRNA-E1, 1 μM crRNA-E2, 1 μM DS-E1, 1 μM DS-E2, 10 μM RP, 0.8 U / μl RNase inhibitor, and 1 × NEBuffer 3.0. ( C ) ISH and H&E staining of corresponding tissue samples. Scale bar = 100 μm for ISH and bar = 40 μm for H&E st aining . The mean FI of 66 clinical samples ( including the EBERs ( + ) group, n = 33, and the EBERs ( -) group, n = 33 ) w as displa y ed in the heat map ( D ) and box plot ( E ) , asterisks indicate t -values ( **** t < 0.0 0 01 ) . ( F ) T he result of SVM learning of normaliz ed FI of 66 samples by CtSDR-based in situ imaging strategy, P means positive, N means negative. The decision surface is described by 4.99 x + 2.78 ( red line ) , the decision surface upper is described by -0.39 ( orange dashed line ) and the decision surface lower is described by -0.71 ( green dashed line ) . ( G ) Efficacy assessment of two methods, CtSDR-based in situ imaging, and ISH. P means positive, N means negative, Sen means sensitivity, and Spe means specificity.
Scheme 1. Illustration of the principle of rapid in situ RNA imaging based on Cas12a thrusting strand displacement reaction.
Rapid in situ RNA imaging based on Cas12a thrusting strand displacement reaction

November 2023

·

84 Reads

·

10 Citations

Nucleic Acids Research

RNA In situ imaging through DNA self-assembly is advantaged in illustrating its structures and functions with high-resolution, while the limited reaction efficiency and time-consuming operation hinder its clinical application. Here, we first proposed a new strand displacement reaction (SDR) model (Cas12a thrusting SDR, CtSDR), in which Cas12a could overcome the inherent reaction limitation and dramatically enhance efficiency through energy replenishment and by-product consumption. The target-initiated CtSDR amplification was established for RNA analysis, with order of magnitude lower limit of detection (LOD) than the Cas13a system. The CtSDR-based RNA in situ imaging strategy was developed to monitor intra-cellular microRNA expression change and delineate the landscape of oncogenic RNA in 66 clinic tissue samples, possessing a clear advantage over classic in situ hybridization (ISH) in terms of operation time (1 h versus 14 h) while showing comparable sensitivity and specificity. This work presents a promising approach to developing advanced molecular diagnostic tools.


Citations (37)


... Multiple factors influence the progression and therapeutic outcomes of tumors, including changes in the immune microenvironment (3)(4)(5)(6), drug resistance (7), and alterations in key signaling pathways, such as PI3K/AKT pathway (8,9). In order to increase patient survival and improve prognosis, current therapeutic options for gliomas include radiation, temozolomide chemotherapy, surgery, and tailored treatment techniques (10). ...

Reference:

DET induces apoptosis and suppresses tumor invasion in glioma cells via PI3K/AKT pathway
Eliminating a barrier: Aiming at VISTA, reversing MDSC-mediated T cell suppression in the tumor microenvironment

Heliyon

... IL11RA interacts with IL11 (84) and can regulate inflammatory responses, bone metabolism, and tumor development through the IL-11 signaling pathway (85). IL11RA is associated with T cells CD8 and can inhibit tumor growth (86), enhance drug resistance (87), and has potential immunomodulatory effects (88), improving survival outcomes (89). Research on the relationship between IL11RA, past tobacco smoking, and NSCLC is limited. ...

Enhancing antitumor immunity and achieving tumor eradication with IL11RA mRNA immunotherapy
  • Citing Article
  • May 2024

International Immunopharmacology

... After being denatured for eight minutes at 98°C using 5× SDS-PAGE buffer, the lysates were separated using 10% SDS-PAGE and then transferred onto PVDF membranes with a pore size of 0.45 µm. Following two hours of blocking with 5% BSA, membranes were incubated with a primary antibody at 4°C for the whole night, followed by an hour incubation at room temperature with a secondary antibody (25). An improved ECL detection kit was used to observe the protein bands, and ImageJ software was used for analysis. ...

Oxytocin alleviates liver fibrosis via hepatic macrophages

JHEP Reports

... [25][26][27] With owning the prots of simplicity, cost-effectiveness, reproducibility and efficiency, CRISPR/ Cas12a systems have offered powerful molecular recognition tools for tracking gene expression in point-of-care diagnostic 28 and live cells. 29 Nevertheless, the existing CRISPR/Cas12a system activation modes are conned to ssDNA or dsDNA targets, and direct recognition of RNA in disease-associated nucleic acid molecules presents signicant challenges. ...

Rapid in situ RNA imaging based on Cas12a thrusting strand displacement reaction

Nucleic Acids Research

... Consequently, several SARS-CoV-2 antibody drugs administrated intranasally are under development, which may allow for faster and better protection against SARS-CoV-2 infection and be more suitable for widespread use in emergency situations. Some NAb nasal spray products have entered the clinical development stage, including the 35B5 mAb nasal spray, which harbors broad neutralization to SARS-CoV-2 variants of concern [25]; A8G6, a combination of two monoclonal NAbs [26]; human IgG1 anti-SARS-CoV-2 antibody cocktail [27]; and SA58, a broad-spectrum anti-SARS-CoV-2 mAb [28,29]. Some of them showed favorable real-world effectiveness in the post-exposure prophylaxis of COVID-19 [26,28,29], offering encouragement for the development of similar drugs. ...

Real-world effectiveness of an intranasal spray A8G6 antibody cocktail in the post-exposure prophylaxis of COVID-19

Signal Transduction and Targeted Therapy

... Tumor cells exploit autophagy to evade immune surveillance through various mechanisms (61,62). Autophagy suppresses apoptosis and the release of antitumor immune factors, such as interferon-g and tumor necrosis factor-a, allowing tumor cells to evade immune detection (63) Moreover, autophagy regulates immune checkpoint molecules, such as PD-L1, on the tumor cell surface, weakening T cell-mediated immune responses. ...

Editorial: Targeting metabolism to activate T cells and enhance the efficacy of checkpoint blockade immunotherapy in solid tumors

... [140] Electrochemical sensors have shown high efficiency, low cost, and ease of operation in detecting nucleic acids. [141] In Figure 9D, Alafeef et al. reported a paper-based electrochemical sensor for detecting viral RNA from SARS-COV2 virus. [142] The sensor used anti-sense ssDNA capped GNPs to capture viral RNA onto a graphene surface, resulting in an increased output voltage. ...

Electrochemical nucleic acid sensors: Competent pathways for mobile molecular diagnostics
  • Citing Article
  • May 2023

Biosensors and Bioelectronics

... Esophageal cancer represents a highly aggressive malignancy within the digestive tract, characterized by a notably elevated propensity for metastasis, and regrettably, patients confronting invasive and metastatic disease frequently experience diminished survival prospects. This somber reality underscores esophageal cancer's standing as a leading contributor to global cancer-related mortality [39][40][41]. Current therapeutic approaches for esophageal cancer encompass a spectrum of modalities, including surgical resection [42], radiation therapy [43], chemotherapy [44], targeted therapy [45], and immunotherapy [46]. ...

Ensemble deep learning enhanced with self-attention for predicting immunotherapeutic responses to cancers

... Primary and secondary drug resistance is a great challenge for cancer therapy [82]. It is of great significance to clarify the sensitivity of each patient to specific drug treatment and improve the response to drug treatment to improve drug efficacy and formulate individualized treatment plans [83]. ...

LRP1B suppresses HCC progression through the NCSTN/PI3K/AKT signaling axis and affects doxorubicin resistance

Genes & Diseases

... Administration of recombinant IL-7 subcutaneously in macaques resulted in dramatic changes within lymph nodes, including enlarged germinal centers, increased IL-21 production, and expansions of B cells and CD4 + and CD8 + T cells (61). However, rIL-7 has thus far only been tested in cancer immune therapy trials in humans (62). It is also possible that the use of liposomal nanoparticles (LNP), as used for the SARS CoV-2 mRNA vaccine in humans (63)(64)(65), which rapidly activates the innate immune system, may bring this type of advantage. ...

IL-7: A promising adjuvant ensuring effective T cell responses and memory in combination with cancer vaccines?