Harbin Medical University
Recent publications
Background Pulmonary hypertension is a pathophysiological condition characterized by multiple forms of regulated cell death. PANoptosis, which is an inflammation‐driven mode of regulated cell death, is regulated by the PANoptosome. Super‐enhancers (SEs) have been implicated in the pathology of pulmonary hypertension by modulating the transcriptional regulation of target genes. However, it remains unclear whether SEs influence the onset of PANoptosis in pulmonary arterial smooth muscle cells (PASMCs) via the regulation of specific target genes. Methods Chromatin immunoprecipitation (ChIP)‐sequencing was integrated with RNA sequencing to identify SE‐regulated target genes in hypoxic PASMCs. Bioinformatics analysis was used to predict transcription factors with potential binding sites in the promoter or SE regions of these target genes. Coimmunoprecipitation and ChIP‐polymerase chain reaction were conducted to validate the epigenetic regulatory mechanisms governing SE‐regulated target genes. Immunoblotting was performed to assess the expression of PANoptosis proteins, whereas YP1 (YO‐PRO‐1; Oxazole yellow) and PI (propidium iodide) fluorescence staining and immunofluorescence were used to evaluate the effects of transcription factors and SE on PANoptosis in PASMCs. Additionally, reverse transcriptase–quantitative polymerase chain reaction, immunoblotting, and ChIP‐polymerase chain reaction assays were used to investigate the impact of the SE region of the target gene on PANoptosis in PASMCs following CRISPR‐Cas9 (clustered regularly interspaced short palindromic repeats‐associated protein 9)‐mediated knockdown of the SE regions of the target genes. Results A combination of ChIP sequencing and RNA sequencing analyses confirmed that FOSL2 (Fos‐like antigen 2) is a target gene regulated by SEs. Bioinformatics predictions revealed that the transcription factor MAZ (myc‐associated zinc finger protein) has binding sites within both the SE and promoter regions of FOSL2 . Coimmunoprecipitation and ChIP‐polymerase chain reaction experiments demonstrated that KAT7 (lysine acetyltransferase 7) interacts with H3K27ac (acetylation of lysine 27 of histone H3) and that circ_chr11_67292179‐67294612 (circ‐myh8) and KAT7 enhances H3K27ac enrichment in the SE region of FOSL2 . CRISPR‐Cas9‐mediated knockdown of the FOSL2 SE resulted in a reduction in PANoptosis in PASMCs. Additionally, ChIP‐polymerase chain reaction assays revealed that FOSL2 functions as a transcription factor that binds to the promoters of PANoptosis‐related genes. Conclusions Circ‐myh8 RNA, which functions jointly with KAT7, enhances H3K27ac enrichment, thereby promoting SE activity for FOSL2 transcription. This process ultimately contributes to the induction of PANoptosis in PASMCs. These findings elucidate the role of the circ‐myh8/KAT7/SE/FOSL2 axis in the regulation of PANoptosis in PASMCs, thus offering new insights into the pathophysiology of PANoptosis in pulmonary hypertension. These findings may lead to novel therapeutic strategies for the treatment of pulmonary hypertension.
Objective: RATIONALE-305 (NCT03777657) demonstrated that tislelizumab plus chemotherapy statistically improved overall survival versus placebo plus chemotherapy as first-line treatment in patients with advanced gastric/gastroesophageal junction adenocarcinoma (GC/GEJC). This analysis examined patient-reported outcomes (PROs) at final analysis. Methods: Adults with previously untreated, unresectable, or metastatic GC/GEJC were randomized (1:1) to tislelizumab or placebo intravenously once every 3 weeks plus chemotherapy. PROs assessed health-related quality of life (HRQoL) using EORTC QLQ-C30 and EORTC QLQ-STO22. A mixed model for repeated measures was used for PRO endpoints at treatment cycles 4 and 6, and time to deterioration was analyzed. Results: Tislelizumab arm had improved outcomes over placebo arm in least-squares (LS) mean change from baseline to cycle 6 for QLQ-C30 global health status/quality of life (GHS/QoL) (LS mean difference, 2.52 [95% CI: 0.29-4.74]), physical functioning (2.46 [0.49-4.43]), fatigue (-3.01 [-5.78 to -0.24]), and STO22 index score (-1.62 [-3.12 to -0.12]) as well as maintenance of upper gastrointestinal symptoms (-1.74 [-3.55-0.06]) and pain/discomfort (-1.88 [-4.03-0.27]). Patients receiving tislelizumab plus chemotherapy had a lower risk for deterioration of GHS/QoL (hazard ratio 0.77 [95% CI: 0.60-0.98]), physical functioning (0.72 [0.57-0.92]), STO22 index score (0.64 [0.45-0.92]), pain/discomfort (0.74 [0.58-0.96]), and upper gastrointestinal symptoms (0.73 [0.56-0.95]). Conclusions: Advanced GC/GEJC patients treated with tislelizumab plus chemotherapy versus placebo plus chemotherapy in first-line had sustained and improved HRQoL. These results, along with previous efficacy and safety data, support tislelizumab plus chemotherapy as a first-line treatment option for GC/GEJC. Trial registration: The RATIONALE-305 trial is registered on ClinicalTrials.gov (ClinicalTrials.gov identifier: NCT03777657).ClinicalTrials.gov identifier: NCT03777657.
Background Brain metastases (BM) remain the leading cause of death in patients with lung adenocarcinoma (LUAD) despite the availability of comprehensive therapeutics. This study aimed to systematically identify crucial genes associated with BM in patients with LUAD, and develop a heuristic search algorithm combining information filtering and multiple stepwise regression to construct a multi-layered transcription factor regulatory network. Methods We used the mRNA expression profiles of primary tumours derived from LUAD patients with different organ metastasis and adjacent normal tissue samples to identify differentially expressed genes specific to BM, and validated it in bulk, spatial and single-cell RNA-sequencing data. Furthermore, we collected 110 stage I–III LUAD specimens to validate the correlation of high PSMD12 (proteasome 26 S subunit, non-ATPase 12) protein expression with BM incidence and patient prognosis. We also developed an in vitro blood–brain barrier (BBB) model to investigate the disruption of PSMD12 on the barrier. Mechanistically, we developed an algorithm to construct an upstream regulator network of PSMD12 overexpression. Results Through integrated bioinformatics analyses, we identified PSMD12 as a critical gene associated with BM. The associations of PSMD12 with BM and poor survival were validated in multiple public datasets comprising 706 patients. Immunohistochemical results confirmed that patients exhibiting high PSMD12 protein expression had a significantly higher incidence of BM within 5 years and shorter progression-free survival. Functional experiments revealed that PSMD12 overexpression in A549 cells disrupted BBB integrity and enhanced tumor cell invasiveness, whereas PSMD12 knockdown attenuated these effects. Furthermore, upstream regulator network and knockdown experiments indicated that downregulated EOMES expression would contribute to PSMD12 overexpression. Conclusions PSMD12 emerges as a crucial gene in promoting BM, potentially serving as an early warning indicator of BM in early-stage LUAD. The upstream regulatory network of PSMD12 overexpression provides potential mechanisms for BM in LUAD patients.
Ischaemic heart disease is an important cause of death in humans, and resupply of blood to damaged myocardium can exacerbate the risk of cardiac I/R injury. Circular RNAs (circRNAs) play an important role in cardiovascular disease. In this study we investigated the regulatory role of circDhx32 in the progression of I/R injury. Cardiac I/R model was established in mice by ligating the left anterior descending coronary artery (LAD) for 45 min, followed by blood reperfusion for 24 h or 2 weeks. For in vitro study, neonatal mouse ventricular cardiomyocytes were subjected to hypoxia-reoxygenation (H/R) assault. CircDhx32 was significantly upregulated in I/R-treated mice and H/R-treated cardiomyocytes. Cardiomyocyte-specific knockdown of circDhx32 ameliorated the pathological outcomes of cardiac I/R injury including improved cardiac function, reduced infarct size and reduced release of cardiac injury biomarkers. The protective effects of circDhx32 silencing were also observed in cardiomyocytes after H/R. We demonstrated that ALKBH5 functioned as an m ⁶ A demethylase, removing the m ⁶ A modification sites of circDhx32. Reduced m ⁶ A modification inhibited recognition and bindin g by the m ⁶ A readers YTHDF2 and YTHDC1, leading to circDhx32 degradation and diminished nucleoplasmic export under pathological conditions. Elevated circDhx32 inhibited the transcriptional activation of AdipoR1 by binding to FOXO1. Conversel y , circDhx32 deficiency alleviated the inflammatory responses in I/R-treated mice and H/R-treated cardiomyocytes including decreased mRNA expression levels and release of inflammatory cytokines such as IL-6, TNF-α and IL-1β potentially through modulation of the AdipoR1-AMPK-NF-κB signaling pathway. In conclusion, ALKBH5 acted as m ⁶ A eraser accompanied by the m ⁶ A readers YTHDF2 and YTHDC1 to promote high expression and nuclear retention of circDhx32 under pathological conditions. CircDhx32 regulated the inflammatory responses to cardiac I/R injury by targeting the AdipoR1-AMPK-NF-κB signaling pathway, which competed with AdipoR1 for FOXO1. These results revea l a novel mechanism underlying cardiac ischaemic injury, and circDhx32 is expected to be a potential therapeutic target for early intervention in ischaemic cardiac disease.
Metabolic dysfunction‐associated steatotic liver disease (MASLD) is a growing health concern worldwide. Aspirin has shown potential in ameliorating MASLD, yet its mechanisms remain incompletely understood. This study aims to investigate the protective effects of aspirin on MASLD by incorporating transcriptomic and metabolomic approaches. Mice were fed a high‐fat diet (HFD) to induce MASLD and treated with aspirin for 12 weeks. Blood and liver samples were collected for biochemical assays, histological analysis, RT‐qPCR, RNA sequencing, and non‐targeted metabolomics. AML12 cells were used for in vitro experiments to validate the findings. Aspirin treatment significantly reduced plasma lipid levels and liver lipid accumulation in HFD‐fed mice. RNA sequencing and non‐targeted metabolomics identified differentially expressed genes (DEGs) and metabolites (DEMs), respectively. These findings were validated through RT‐qPCR for the DEGs and targeted mass spectrometry for the DEMs. Enrichment analyses highlighted several key pathways, including lipid metabolism, PPAR signaling, and bile acid metabolism. Integrated transcriptomic and metabolomic analysis identified 42 overlapped pathways that may mediate the protective effects of aspirin. In vitro experiments confirmed that aspirin reduced lipid accumulation and inflammation in palmitic acid‐treated AML12 cells. Molecular docking confirmed strong binding between aspirin/cholic acid and SULT2A3. SULT2A3 was upregulated in MASLD patients and HFD‐fed mice. Functional studies revealed SULT2A3 overexpression exacerbated PA‐induced lipid accumulation, inflammation, and bile acid dysregulation, whereas its knockdown or aspirin treatment mitigated these effects. Aspirin ameliorates MASLD by modulating SULT2A3‐mediated bile acid metabolism and inflammatory pathways. Sult2a3 emerges as a potential target for the treatment of MASLD.
Background Hospital workplace violence (WPV) is a serious obstacle to the sustainable development of global health care, Therefore, it is necessary to adjust and optimize the current WPV prevention and control policies (WPVPCPs), and policy evaluation is the first prerequisite of this link. However, there is little literature on WPVPCPs. The aim of our study is to quantitatively evaluate the China’s WPVPCPs, and provide empirical evidence for the formulation, adjustment and optimization of relevant policies. Methods On the basis of the PMC index model, text mining technology was used to construct a WPVPCPs evaluation system. Overall, 37 WPVPCPs published in China were quantitatively evaluated, and representative policies were selected to draw PMC-Surface chart. Results Among 37 WPVPCPs, 1 was excellent, 27 were good and 9 were acceptable. The average PMC index was 6.43. Conclusions The overall quality of WPVPCPs is high, but there is still room for improvement. Specifically, the overall score of primary variables such as policy perspective and policy level is low, and the score of secondary variables of policy nature, policy object, policy tool and policy prescription is unbalanced. These aspects may be the entry point for effective improvement of WPV, so they should be taken seriously in the development and optimization of relevant policies in the future.
Background Temozolomide (TMZ) is an important chemotherapeutic agent for glioma treatment. However, the emergence of drug resistance impedes its application. Traditional population-level studies are limited in elucidating resistance mechanisms. Advances in single-cell and spatial transcriptomics technologies provide feasible resolution for studying the cellular composition and dynamics of tumors. In this study, we investigated the heterogeneity of gliomas associated with TMZ resistance at the single-cell and spatial transcriptome levels to identify the resistance mechanisms and potential therapeutic strategies. Methods Single cell sequencing technology was utilized to identify the cellular clusters of gliomas. Drug perturbation analysis and cellular propensity analysis revealed key cluster responding to TMZ. Enrichment analysis was preformed to explore the function of clusters. Transcription factor activity analysis revealed key transcription factors contributing to tumor resistance. Spatial transcriptome data and bulk RNA-seq data validates the role of key transcription factors. Downstream targets of key transcription factors were predicted and validated using gene regulation assays. Drug sensitivity analyses were used to seek viable strategies to overcome drug resistance. Results Glioma cells from before and after temozolomide treatment samples were classified into six clusters: NPC-like cluster, OPC-like cluster, MES-like cluster, AC-like cluster, OC, and Neuron. NPC-like clusters exhibited strong stemness and DNA repair capacity. The activity of MAZ in NPC-like cluster was significantly enhanced after TMZ treatment. The proportion of MAZ( +)_NPC-like cluster was higher in TMZ treated samples. Patients with high proportion of MAZ( +)_NPC-like cluster had poorer survival. Upregulation of MAZ is able to enhance drug resistance in glioma cells, but this phenomenon disappeared when FoxM1 expression was further silenced. The combination of paclitaxel and Trametinib is a promising strategy to overcome resistance. Conclusions NPC-like cluster is prevalent in recurrent and drug-resistant gliomas. MAZ transcription factors are critical regulators that promote the development of drug resistance in NPC-like clusters by enhancing the capacity of DNA repair and stemness. Patients with high proportions of MAZ( +)_NPC-like clusters have poor TMZ sensitivity and prognosis. MAZ enhances stemness and drug resistance in glioma cells by upregulating FOXM1 expression. The combination of paclitaxel and paclitaxel is a promising therapeutic strategy for treating gliomas and overcoming drug resistance.
Background Plasma cell-free DNA (cfDNA) methylation is emerging as a non-invasive marker for various cancers. We aimed to identify specific methylation markers for diagnosis, differentiation from benign tumors, and prognosis of breast cancer (BC), which are essential for clinical decision-making yet seldom examined together. Methods BC-specific methylation markers were identified using an in-house 850K dataset combined with large-scale publicly available 450 or 850K datasets. Multiplex digital droplet PCR (mddPCR) assays were developed to detect methylation in cfDNA from 201 BC patients, 83 healthy donors, and 71 individuals harboring benign tumors. Diagnostic and prognostic performance were evaluated using logistic and Cox regression models, respectively. The basic mechanism of a selected gene was explored through in vitro experiments. Results We identified 21 BC-specific methylated CpG sites that distinguished BC from tumor-adjacent tissues with high diagnostic accuracy. In the cfDNA cohort, three mddPCR assays targeting eight markers achieved an area under the curve (AUC) of 0.856 (95% CI = 0.814–0.898) for distinguishing BC from healthy controls, and 0.742 (95% CI = 0.684–0.801) for differentiating BC from benign tumors. Notably, combining these methylation markers with mammography and ultrasound improved diagnostic performance, resulting in an AUC of 0.898 (95% CI = 0.858–0.938) for differentiating BC from benign tumors. In the TCGA-BC dataset, prognostic model based on six sites was associated with poor overall survival prognosis (hazard ratio = 2.826, 95%CI: 1.841–4.338, p < 0.0001). In vitro experiments elucidated that FAM126A overexpression regulates BC cells malignant phenotypes. Conclusions Our study demonstrated potential values of methylation-based markers in the detection and prognosis of BC.
Transcriptome sequencing has become essential in clinical tumor research, providing in-depth insights into the biology and functionality of tumor cells. However, the vast amount of data generated and the complex relationships between gene expressions make it challenging to effectively identify clinically relevant information. In this study, we developed a method called Gene Swin Transformer to address these challenges. This approach converts transcriptomic data into Synthetic Image Elements (SIEs). We utilized data from 12 datasets, including GSE17536-GSE103479 datasets (n = 1771) and The Cancer Genome Atlas (n = 459), to generate SIEs. These elements were then classified based on survival time using deep learning algorithms to predict colorectal cancer prognosis and build a reliable prognostic model. We trained and evaluated four deep learning models—BeiT, ResNet, Swin Transformer, and ViT Transformer—and compared their performance. The enhanced Swin-T model outperformed the other models, achieving weighted precision, recall, and F1 scores of 0.708, 0.692, and 0.705, respectively, along with area under the curve values of 80.2%, 72.7%, and 76.9% across three datasets. This model demonstrated the strongest prognostic prediction capabilities among those evaluated. Additionally, the PEX10 gene was identified as a key prognostic marker through both visual attention matrix analysis and bioinformatics methods. Our study demonstrates that the Gene Swin model effectively transforms Ribonucleic Acid (RNA) sequencing data into SIEs, enabling prognosis prediction through attention-based algorithms. This approach supports the development of a data-driven, unified, and automated model, offering a robust tool for classification and prediction tasks using RNA sequencing data. This advancement presents a novel clinical strategy for cancer treatment and prognosis forecasting.
Intrahepatic cholangiocarcinoma (iCCA) represents a growing health concern due to its increasing incidence and poor prognosis, highlighting the urgent need for biomarkers and therapeutic targets. This study utilized BayesPrism deconvolution, Weighted Gene Co-expression Network Analysis (WGCNA), and Summary Mendelian Randomization (SMR), integrated with single-cell RNA sequencing (scRNA-seq) data, to analyze the tumor microenvironment. Seven distinct cell subpopulations, including Exhausted CD8 + T cells (Tex), were identified. Notably, scPagwas analysis revealed gene enrichment in UQCRH, HINT1, and AKR1C3, associated with Tex. BayesPrism analysis confirmed their increased presence in the tumor microenvironment, indicating a role in immune evasion. WGCNA identified 594 genes linked to these cells, with PNO1 and AKR1C5P emerging as potential disease-associated genes. These findings highlight the critical role of Tex in immune suppression and identify key genes for further investigation in iCCA progression and treatment strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-02970-w.
Background & Aims Hepatocellular carcinoma (HCC) remains a leading cause of cancer mortality in China. Anti-angiogenic drugs (AADs) plus immune checkpoint blockade (ICB) combination therapy shows considerable promise for HCC; however, its efficacy is hampered by immunosuppression within the tumor microenvironment. High endothelial venules (HEVs) facilitate lymphocyte migration and tumor infiltration. The aim was to study the formation, functional mechanisms, and clinical relevance of HEVs in the treatment of HCC with combination therapy. Approach & Results Single-nucleus RNA sequencing, flow cytometry, and immunohistochemistry revealed increased HEV expression and higher CD3⁺ T-cell infiltration in HCC tissue after combination AAD and ICB therapy. Multiplex immunohistochemistry and spatial analysis demonstrated that CCR7⁺CD8⁺ T cells were spatially associated with HEVs. Pseudotime analysis of human T cells and treatment of Hepa1-6 orthotopic liver tumor mouse models with CCR7 ⁺ CD8 ⁺ T-cell transfusions were used to show that CCR7 ⁺ CD8 ⁺ T cells can differentiate into cytotoxic effector T cells. The same models demonstrated that combination therapy activated VEGFC and non-canonical NF-κB pathways, promoting HEV formation. Kaplan-Meier analysis revealed that high HEV density correlated with improved clinical response and prolonged survival. Conclusions HEVs are pivotal in modulating immune activity within the HCC tumor microenvironment. Targeting the VEGFC–NF-κB (non- canonical)–HEV axis could be a promising therapeutic strategy to enhance antitumor immunity and improve outcomes in patients with HCC who are receiving combination AAD plus ICB therapy.
Institution pages aggregate content on ResearchGate related to an institution. The members listed on this page have self-identified as being affiliated with this institution. Publications listed on this page were identified by our algorithms as relating to this institution. This page was not created or approved by the institution. If you represent an institution and have questions about these pages or wish to report inaccurate content, you can contact us here.
1,371 members
Yongsheng Kevin Li
  • School of Interdisciplinary Medicine and Engineering
Yong ji
  • State Key Laboratory of Frigid Zone Cardiovascular Diseases
Shaojun Zhang
  • Department of Bioinformatics
Liming Zhang
  • Department of Neurology
Xiaoping Ren
  • Hand and Microsurgical Center/Departmnt of Orthopeadic
Information
Address
Harbin, China