Xingmin Chen’s research while affiliated with University of Electronic Science and Technology of China 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 (2)


Origins of CAFs. CAFs may originate from various cell types including pericytes, adipocytes, stellate cells, mesothelial cells, epithelial cells, endothelial cells, mesenchymal Stem Cells (MSCs) and normal fibroblast. This figure was created with BioRender.com
Methodology for studying CAFs. (A) CAFs may be obtained through primary culture, or transformation from other cells, such as normal fibroblasts or pancreatic stellate cells (PSCs), for further researches. (B) Animal models of CAFs include patient-derived xenografts (PDX) and transplanted tumor models established from cell lines. Genetically engineered mouse models (GEMMs) can spontaneously develop tumors, better simulating the interaction between CAFs and TME. These genetically engineered mice also allow for the specific elimination of CAF subsets using drugs such as diphtheria toxin, as well as lineage tracing of CAF subsets through the expression of specific markers. (C) In vitro approaches to study CAF-TME interactions include 2D/3D co-culture, conditioned media transfer, and tumor organoid models. In vivo methods involve co-inoculation of CAFs and tumor cells. (D) CAF research integrates techniques such as western blotting (WB), flow cytometry, and immunofluorescence. Advanced tools like single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics provide deeper functional insights. This figure was created with BioRender.com
CAFs modulate ECM to promote tumor growth. (A) CAFs promote the deposition of extracellular matrix, forming a physical barrier that hinders immune cell infiltration and drug penetration, thereby creating obstacles for anti-tumor treatment. (B) CAFs remodel ECM, facilitating tumor cell invasion and metastasis. (C) CAFs regulate the balance between dense ECM and loose ECM, thus promoting cancer progression. This figure was created with BioRender.com
CAFs modulate the tumor immune microenvironment (TIME). (A) CAFs inhibit the function of effector T cells by secreting IL-6, CXCL12, TGF-β, and by expressing PD-L1; they also recruit regulatory T cells (Tregs). (B) CAFs recruit macrophages through cytokines such as IL-6, CXCL12, and IL-33, and promote M2 polarization and inducing immunosuppresive subses like lipid-associated macrophages (LAMs). (C) CAFs promote N2 polarization of TANs, upregulate PD-L1, and stimulate neutrophil extracellular trap (NET) formation. (D) CAFs enhance infiltration of monocytic (M-MDSCs) and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) via tumor-derived CSF-1. This figure was created with BioRender.com
Interplays between CAFs and tumor cells. (A) CAFs fuel tumor cell glycolysis via CCL6 and CCL12 secretion; In addition, Abca8a + CAFs facilitate tumor cell growth by secreting lipid droplets. (B) CAFs enhance tumor proliferation and therapy resistance through IL-6, HGF, and FGF2 secretion, and via exosomal long non-coding RNAs (lncRNAs). (C) CAFs induce epithelial-mesenchymal transition (EMT) via Hedgehog ligands, TGF-β, and HGF. (D) CAF-derived exosomes containing miR-522 suppress lipid peroxidation to inhibit ferroptosis. (E) Complement component 5a (C5a) activates GPR77 + CAFs, inducing tumor stemness via IL-6, IL-8, and TGF-β. This figure was created with BioRender.com

+1

Cancer associated fibroblasts in cancer development and therapy
  • Literature Review
  • Full-text available

March 2025

·

45 Reads

·

2 Citations

Journal of Hematology & Oncology

Hongyuan Jia

·

Xingmin Chen

·

Linling Zhang

·

Meihua Chen

Cancer-associated fibroblasts (CAFs) are key players in cancer development and therapy, and they exhibit multifaceted roles in the tumor microenvironment (TME). From their diverse cellular origins, CAFs undergo phenotypic and functional transformation upon interacting with tumor cells and their presence can adversely influence treatment outcomes and the severity of the cancer. Emerging evidence from single-cell RNA sequencing (scRNA-seq) studies have highlighted the heterogeneity and plasticity of CAFs, with subtypes identifiable through distinct gene expression profiles and functional properties. CAFs influence cancer development through multiple mechanisms, including regulation of extracellular matrix (ECM) remodeling, direct promotion of tumor growth through provision of metabolic support, promoting epithelial-mesenchymal transition (EMT) to enhance cancer invasiveness and growth, as well as stimulating cancer stem cell properties within the tumor. Moreover, CAFs can induce an immunosuppressive TME and contribute to therapeutic resistance. In this review, we summarize the fundamental knowledge and recent advances regarding CAFs, focusing on their sophisticated roles in cancer development and potential as therapeutic targets. We discuss various strategies to target CAFs, including ECM modulation, direct elimination, interruption of CAF-TME crosstalk, and CAF normalization, as approaches to developing more effective treatments. An improved understanding of the complex interplay between CAFs and TME is crucial for developing new and effective targeted therapies for cancer. Graphical abstract

Download

Figure 2. Barriers of PDT in treating cancer. (A) At the host level, ① PDT affects the cytoskeleton and cell adhesion, potentially enhancing cancer cell migration and invasion. ② Inadequate immune surveillance and regulation can limit the efficacy of PDT against distant lesions. (B) At the TME level, ① the hypoxic TME re-duces oxygen availability and stabilizes hypoxia-inducible factor 1 (HIF-1). ② Damage to blood vessels impairs oxygen supply, exacerbating hypoxia. ③ The location and shape of tumors can affect the absorption, distribution, and excitation of PSs, reducing ROS production. (C) At the subcellular level, ① Up-regulation of transport proteins excrete PSs. ② Enhanced antioxidant systems scavenge ROS in tumor tissues. ③ Activation of pro-survival signaling pathways promotes transcription and tumor progression. ④ Up-regulation of heat shock proteins facilitates the repair of proteins damaged by ROS.
Figure 3. Examples of NGs and PDT combined applications. (A) Human motion driven self-powered photodynamic system for long-term autonomous cancer therapy [36]. (B) A self-powered wireless detachable drug/light injector for metronomic photodynamic therapy in cancer treatment [146]. (C) Implanted, wireless, self-powered photodynamic therapeutic tablet synergizes with ferroptosis inducer for effective cancer treatment [147]. (D) Implantable self-powered therapeutic pellet for wireless photodynamic/sonodynamic hybrid therapy of cancer recurrence inhibition and tumor regression [148]. (E) Self-powered, implantable, and wirelessly controlled NO generation system for intracranial neuroglioma therapy [13]. (A) Reprinted with permission from [36]. Copyright 2020 American Chemical Society. (B) Reprinted with permission, LN: 5882360170575. (D) Reprinted with permission, LN: 5882810859864. (E) Reprinted with permission, LN: 5882811413873.
Figure 5. Strategies employed by NGs to overcome tumor resistance to PDT. (A) At the host level, NG-supported devices detect various biomarkers for early identification of metastasis and enhance immune activity to improve survival. (B) At the tumor level, ①A TENG-based sensor detect dopamine levels with anti-angiogenic properties. ② PENG/TENG systems facilitate thermal therapy, increasing O2 supply, relieving hypoxia, and enhancing ROS production. RR-TENG or TENG-Cat directly stimulate ROS production. ③ PENG/TENG-supported drug delivery systems(DDSs) enhance PS absorption, excitation, and ROS production for deep-seated or irregularly shaped tumors. (C) At the subcellular level, ① NG devices influence cell death signals, with TENG enhancing ROS production and inducing necrosis or enhancing apoptosis through Bcl-2 gene silencing or autophagy suppression. ② NG devices suppress pro-survival signals by increasing ROS production and silencing pro-survival genes or activating the AMPK signal to regulate mTOR-HIF1 signaling. ③Silencing the Mutt homolog 1(MTH1) gene reduces protein repair damaged by ROS. ④ TENG devices silence P-gp and ABCG2 to reduce photosensitizer exocytosis. TENG-based glucose sensors detect ATP production. ⑤ A designed D-TENG enhances cancer cell endocytosis and PS absorption.
Overcoming Barriers in Photodynamic Therapy Harnessing Nanogenerators Strategies

October 2024

·

10 Reads

·

2 Citations

International Journal of Biological Sciences

Yi Zhou

·

Pingjin Zou

·

Xingmin Chen

·

[...]

·

Meihua Chen

Photodynamic therapy (PDT) represents a targeted approach for cancer treatment that employs light and photosensitizers (PSs) to induce the generation of reactive oxygen species (ROS). However, PDT faces obstacles including insufficient PS localization, limited light penetration, and treatment resistance. A potential solution lies in nanogenerators (NGs), which function as self-powered systems capable of generating electrical energy. Recent progress in piezoelectric and triboelectric NGs showcases promising applications in cancer research and drug delivery. Integration of NGs with PDT holds the promise of enhancing treatment efficacy by ensuring sustained PS illumination, enabling direct electrical control of cancer cells, and facilitating improved drug administration. This comprehensive review aims to augment our comprehension of PDT principles, explore associated challenges, and underscore the transformative capacity of NGs in conjunction with PDT. By harnessing NG technology alongside PDT, significant advancement in cancer treatment can be realized. Herein, we present the principal findings and conclusions of this study, offering valuable insights into the integration of NGs to overcome barriers in PDT.

Citations (1)


... In turn, the excited photosensitizer can react with molecular oxygen to form singlet oxygen ( 1 O 2 ) through energy transfer (type II reaction) [148]. On the other hand, ROS generated during PDT exerts cytotoxic effects by the cellular degradation of lipids, proteins, and DNA, whereby the plasma membrane and organelles such as mitochondria, endoplasmic reticulum (ER), Golgi apparatus, and lysosomes are particularly susceptible to ROS [149]. Further disruption of the integrity and function of biomembranes is caused by photodynamic reaction between 1 O 2 and unsaturated lipids, contributing to the formation of lipid peroxides [149]. ...

Reference:

Phytochemical-based nanosystems: recent advances and emerging application in antiviral photodynamic therapy
Overcoming Barriers in Photodynamic Therapy Harnessing Nanogenerators Strategies

International Journal of Biological Sciences