1
|
Xie J, Zheng Z, Wang B, Zhang J, Jiang J, Wu F, Zhong X, Chen J. LncRNA HOTAIR promotes aerobic glycolysis by recruiting Lin28 to induce inflammation and apoptosis in acute lung injury. RNA Biol 2025; 22:1-12. [PMID: 40052944 PMCID: PMC11901367 DOI: 10.1080/15476286.2025.2475255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition with high rates of morbidity and mortality. Recently, there has been growing evidence suggesting a link between lncRNA HOTAIR and ALI. Nonetheless, the precise role and mechanism of lncRNA HOTAIR in ALI remain to be fully elucidated. siHOTAIR transfection, qPCR detection (HOTAIR), ELISA (TNF-α, IL-6, and IL-1β), Lactate detection, Glucose uptake experiment, Cell Apoptosis Analysis, Fluorescence in situ hybridization (FISH) assay. Through siHOTAIR transfection, we discovered that HOTAIR plays a role in the secretion of inflammatory factors in ALI and further regulates glucose uptake and metabolism in lung epithelial cells. Moreover, a comparison between HOTAIR knockdown cells and HOTAIR overexpression cells revealed that HOTAIR promotes cellular aerobic sugar metabolism, leading to increased secretion of inflammatory factors and cell apoptosis. Our in-depth research also identified an interaction between HOTAIR and the LIN28 protein. Knocking down HOTAIR resulted in the downregulation of LIN28 protein expression, which subsequently inhibited the expression of the glucose transporter GLUT1. This indicates that HOTAIR facilitates glucose uptake and boosts cellular aerobic glycolysis by modulating the LIN28 protein, thereby promoting inflammation and apoptosis in acute lung injury. The research findings presented in this article offer significant insights into the function of HOTAIR in ALI and suggest a potential therapeutic target for the treatment of this condition.
Collapse
Affiliation(s)
- Junjie Xie
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Zhicong Zheng
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianfang Zhang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Junqi Jiang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Fengde Wu
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Xiangming Zhong
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Jianfeng Chen
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Wei L, Wang H, Ye X, Yue J, Guo H, Mao D, Li X, Sun Y, Liu C, Liu Y, Chen Y. Oxymatrine and astragaloside IV co-loaded liposomes: Scale-up purposes and their enhancement of anti-PD-1 efficacy against breast cancer. Mater Today Bio 2025; 32:101634. [PMID: 40177381 PMCID: PMC11964553 DOI: 10.1016/j.mtbio.2025.101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 04/05/2025] Open
Abstract
The response rate of programmed cell death protein-1 (PD-1) inhibitors in breast cancer remains unsatisfactory, primarily due to the limited infiltration and activity of tumor-infiltrating T lymphocytes (TILs). Previous studies demonstrated that oxymatrine (Om) and astragaloside IV (As) could enhance TIL infiltration and function by inhibiting cancer-associated fibroblasts (CAFs) and promoting mitochondrial activity in TILs, respectively. Thus, combining Om and As may be a promising strategy to improve the antitumor effects of PD-1 inhibitors in breast cancer. However, co-delivery above drugs into breast cancer tissue is challenging due to their low bioavailability and distinct physicochemical properties. This study addresses this challenge by formulating Om and As co-loaded liposomes (Om-As-Lip) and comparing the scale-up production methods: high-pressure homogenization (EP-HPH) and microfluidics. Om-As-Lip prepared via microfluidics demonstrated superior entrapment efficiency (As: 99.03 ± 0.04 %, Om: 67.01 ± 0.02 %) and a significantly higher production rate (22.12 mL/min) compared to EP-HPH (1.19 mL/min). Additionally, Om-As-Lip produced by microfluidics increased the area under the curve (AUC) (Om: 6.17-fold, As: 2.07-fold) and maximum concentration (Cmax) (Om: 1.58-fold, As: 3.49-fold) compared to the free drugs. Importantly, Om-As-Lip enhanced the antitumor efficacy of α-PD-1 by inhibiting CAF activation and boosting TIL activity, resulting in a tumor inhibition rate of 61.2 % and extended survival in mice. This work presents a novel perspective for scaling up co-delivered formulations of drugs with differing polarities to improve breast cancer immunotherapy.
Collapse
Affiliation(s)
- Liangyin Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Hong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xietao Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Junfan Yue
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Hong Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Dengxuan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xia Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yeyang Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
3
|
Aloliqi AA, Alnuqaydan AM, Albutti A, Alharbi BF, Rahmani AH, Khan AA. Current updates regarding biogenesis, functions and dysregulation of microRNAs in cancer: Innovative approaches for detection using CRISPR/Cas13‑based platforms (Review). Int J Mol Med 2025; 55:90. [PMID: 40242952 PMCID: PMC12021393 DOI: 10.3892/ijmm.2025.5531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
MicroRNAs (miRNAs) are short non‑coding RNAs, which perform a key role in cellular differentiation and development. Most human diseases, particularly cancer, are linked to miRNA functional dysregulation implicated in the expression of tumor‑suppressive or oncogenic targets. Cancer hallmarks such as continued proliferative signaling, dodging growth suppressors, invasion and metastasis, triggering angiogenesis, and avoiding cell death have all been demonstrated to be affected by dysregulated miRNAs. Thus, for the treatment of different cancer types, the detection and quantification of this type of RNA is significant. The classical and current methods of RNA detection, including northern blotting, reverse transcription‑quantitative PCR, rolling circle amplification and next‑generation sequencing, may be effective but differ in efficiency and accuracy. Furthermore, these approaches are expensive, and require special instrumentation and expertise. Thus, researchers are constantly looking for more innovative approaches for miRNA detection, which can be advantageous in all aspects. In this regard, an RNA manipulation tool known as the CRISPR and CRISPR‑associated sequence 13 (CRISPR/Cas13) system has been found to be more advantageous in miRNA detection. The Cas13‑based miRNA detection approach is cost effective and requires no special instrumentation or expertise. However, more research and validation are required to confirm the growing body of CRISPR/Cas13‑based research that has identified miRNAs as possible cancer biomarkers for diagnosis and prognosis, and as targets for treatment. In the present review, current updates regarding miRNA biogenesis, structural and functional aspects, and miRNA dysregulation during cancer are described. In addition, novel approaches using the CRISPR/Cas13 system as a next‑generation tool for miRNA detection are discussed. Furthermore, challenges and prospects of CRISPR/Cas13‑based miRNA detection approaches are described.
Collapse
Affiliation(s)
- Abdulaziz A. Aloliqi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Abdullah M. Alnuqaydan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Aqel Albutti
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Basmah F. Alharbi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| |
Collapse
|
4
|
Lu J, Zhou Y, Song YX, Wang JY, Xian JX. Natural alkaloids modulating macrophage polarization: Innovative therapeutic strategies for inflammatory, cardiovascular, and cancerous diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156709. [PMID: 40250001 DOI: 10.1016/j.phymed.2025.156709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/21/2025] [Accepted: 03/29/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Macrophage polarization, switching between pro-inflammatory M1 and anti-inflammatory M2 states, is crucial for disease dynamics in inflammatory, metabolic, and cancer contexts. Modulating this polarization is a clinical challenge, but natural alkaloids, with their potent anti-inflammatory and immunomodulatory effects, show promise in reprogramming macrophage phenotypes. PURPOSE This review explores the applications of natural alkaloids-such as matrine, berberine, koumine, sophoridine, and curcumin-in modulating macrophage polarization. It aims to highlight their potential in reprogramming macrophage phenotypes and improving therapeutic outcomes across various diseases. METHODS A comprehensive literature review was conducted using databases like PubMed, Web of Science, Science Direct and Google Scholar, employing targeted keywords related to natural alkaloids, macrophage polarization, and disease treatment. The analysis primarily focused on articles published between 2020 and 2024. RESULTS This review summarizes how natural alkaloids regulate macrophage polarization, promoting the M2 phenotype to reduce inflammation, thereby playing a therapeutic role in anti-inflammatory, cardiovascular, and metabolic diseases. At the same time, they also promote M1 polarization to inhibit tumor development. CONCLUSION Accumulating evidence demonstrates that macrophage polarization regulation by natural alkaloids holds notable clinical value for disease intervention. They alleviate inflammation, enhance antitumor immunity, and improve treatment outcomes, demonstrating their importance in innovative therapeutic strategies. Moreover, combining alkaloids with immunotherapy enhances treatment efficacy, further highlighting their versatility in a variety of therapeutic applications.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ying Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi-Xuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie-Ying Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jia-Xun Xian
- Traditional Chinese Medicine Hospital of Meishan, Meishan 620010, China.
| |
Collapse
|
5
|
Yin S, Wang J, Jia Y, Wang X, Zhao Y, Liu T, Lv W, Duan Y, Zhao S, Wang S, Liu L. Sleep deprivation-induced sympathetic activation promotes pro-tumoral macrophage phenotype via the ADRB2/KLF4 pathway to facilitate NSCLC metastasis. iScience 2025; 28:112321. [PMID: 40276761 PMCID: PMC12018092 DOI: 10.1016/j.isci.2025.112321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/01/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Sleep deprivation is one of concomitant symptoms of cancer patients, particularly those with non-small cell lung cancer (NSCLC). The potential effect of sleep deprivation on tumor progression and underlying mechanisms remain to be fully investigated. Using a sleep-deprived tumor-bearing mouse model, we found that sleep deprivation altered immune cell composition and regulated pro-tumoral M2 macrophage polarization by the sympathetic nervous system. Furthermore, we identified a role of catecholaminergic neurons in the rostral ventrolateral medulla (RVLM) in influencing NSCLC metastasis. Clinical analyses revealed a correlation between sympathetic-related indicators and poor prognosis. Mechanistically, our findings indicate that sleep deprivation facilitates the polarization of pro-tumoral macrophages by upregulating β2-adrenergic receptor (ADRB2), which subsequently enhances the expression of Kruppel-like transcription factor 4 (KLF4) through the JAK1/STAT6 phosphorylation pathway. These findings highlight a neuro-immune mechanism linking sleep deprivation to NSCLC metastasis, suggesting that targeting the ADRB2/KLF4 axis could improve outcomes for sleep-deprived NSCLC patients.
Collapse
Affiliation(s)
- Shuxian Yin
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
- Hebei Key Laboratory of Stomatology, Hebei Technology Innovation Center of Oral Health, Hebei Medical University, Shijiazhuang, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Xiaoyi Wang
- Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, China
| | - Yan Zhao
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Tianxu Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Wei Lv
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Song Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Sheng Wang
- Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
- International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
6
|
Lee MS, Thomas SM, Louie AD, Rosenberger LH, Bansal R, DiLalla G, Parrish KJM, Wang T, Woriax HE, Hwang ES, Plichta JK. Comparison of survival outcomes for patients with Stage III vs de novo Stage IV breast cancer. Cancer 2025; 131:e35891. [PMID: 40327594 DOI: 10.1002/cncr.35891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/23/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE Improvements in systemic therapy have resulted in significant heterogeneity in survival outcomes for metastatic breast cancer patients. As such, recently proposed staging guidelines for de novo metastatic breast cancer stratify patients into four categories (IVA/IVB/IVC/IVD). Expanding on this, overall survival (OS) outcomes for patients with Stage III vs Stage IV breast cancer were compared based on the previously defined American Joint Committee on Cancer guidelines and recently proposed subgroups for de novo metastatic breast cancer. METHODS Adult patients diagnosed with Stage III or IV breast cancer in the National Cancer Database (2010-2019) were stratified as IIIA/B/C (American Joint Committee on Cancer, 8th edition) or IVA/B/C/D. OS was estimated using the Kaplan-Meier method. Cox proportional hazards models estimated the association between stage subgroups and OS. RESULTS Among 81,128 patients (median follow-up, 76.8 months), 83.5% were Stage III and 16.5% Stage IV. Unadjusted 3-year OS rates were 85.7% for Stage III versus 68.3% for Stage IV. From Stage III to Stage IV, OS declined but there was notable convergence in OS between subgroups. The unadjusted 3-year OS for IIIC was 69.6%, which was lower than IVA (87.0%) and IVB (78.4%). Adjusted analysis showed similar trends, with the HR for IIIC at 1.94, which was worse than IVA at 1.20 and IVB at 1.83 (ref: IIIA, overall p < .001). CONCLUSIONS It was demonstrated that the survival outcomes for select patients with Stage IV breast cancer have significant convergence in OS with some patients with Stage III disease. These findings may be important for patient counseling, treatment approaches, and clinical trial design.
Collapse
Affiliation(s)
- Michael S Lee
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Samantha M Thomas
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Anna D Louie
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Laura H Rosenberger
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Rani Bansal
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Gayle DiLalla
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Kendra J M Parrish
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Ton Wang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Hannah E Woriax
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Jennifer K Plichta
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Department of Population Health Sciences, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
7
|
Xu K, Kang Y, Wang J, Hou Y, Zheng W, Tian W, Liang C, Liu Y, Xiang X. SYT7 accelerates nasopharyngeal carcinoma progression via ALDH1A3-mediated STAT3 signaling activation. Oncogenesis 2025; 14:16. [PMID: 40346036 DOI: 10.1038/s41389-025-00558-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a special histological and ethical type of head and neck cancer with unsatisfactory clinical outcome. Thus, exploring effective molecular targets is critical for NPC treatment. We observed increased expression levels of synaptotagmin-7 (SYT7) in NPC tissues, which correlated with unfavorable prognoses. Furthermore, knockdown of SYT7 in NPC cells suppressed proliferation and migration rates, and enhanced apoptosis. In contrast, overexpression of SYT7 accelerated NPC tumor growth. Using whole-genome gene arrays and immunoprecipitation-mass spectrometry assays, aldehyde dehydrogenase 1 family member A3 (ALDH1A3), a regulator of glycolytic metabolism, was identified as a critical downstream target of SYT7. Mechanistically, SYT7 binds and promotes ALDH1A3 deubiquitination, resulting in decreased ALDH1A3 degradation. Notably, we also observed an increased expression of ALDH1A3 in NPC. More importantly, the knockdown of ALDH1A3 resulted in suppressed proliferation, migration, glycolysis, and promoted apoptosis, all of which could be restored by the overexpression of SYT7 in NPC cells. Taken together, we found that SYT7 increases ALDH1A3-mediated STAT3 activation and glycolysis, contributing to NPC progression, which provides a possible molecular mechanism for the development of targeted therapeutics interventions.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Kang
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Otolaryngology, Zibo Central Hospital, Zibo, China
| | - Ying Hou
- Department of Otolaryngology, Zibo Central Hospital, Zibo, China
| | - Wenxiang Zheng
- Central of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Wenxiu Tian
- Central of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Chuanjie Liang
- Central of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Yongliang Liu
- Department of Otolaryngology, Zibo Central Hospital, Zibo, China.
| | - Xinxin Xiang
- Central of Translational Medicine, Zibo Central Hospital, Zibo, China.
| |
Collapse
|
8
|
Chen J, Zhao S, Yan H, Huang Y, Wei C, Liu J, Sun J. Plasma SARS-CoV-2 nucleocapsid antigen levels are associated with lung infection and tissue-damage biomarkers. Virus Res 2025:199580. [PMID: 40339608 DOI: 10.1016/j.virusres.2025.199580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND SARS-CoV-2 nucleocapsid (N) antigen has been confirmed in the peripheral blood of patients with new coronavirus infection,yet its diagnostic and prognostic significance remains unclear. This study aimed to characterize the dynamics of SARS-CoV-2 N antigenemia in patients with novel coronavirus positivity, and to assess its potential association with clinical severity and plasma biomarker levels. METHODS We analyzed the level of SARS-CoV-2 N antigen, spike receptor-binding domain (S-RBD) IgG, neutralizing antibodies (NAb) and tissue-damage biomarkers was assessed in 180 plasma samples from 51 SARS-CoV-2-positive individuals. Plasma antigen levels were compared with concurrent respiratory nucleic acid amplification test results. RESULTS Patients with Ct values below 30 showed significantly different serum antigen levels compared to those with Ct values above 30 (p < 0.01). However, no significant positive correlation was found between respiratory viral load and serum antigen levels. Further analysis revealed that patients with pneumonia had markedly higher serum antigen levels than those without (p < 0.0001). Additionally, serum amyloid A (SAA) and ferritin (Fe) levels were significantly elevated in the antigenemia-positive group compared to the negative group, while procalcitonin (PCT) and interleukin-6 (IL-6) levels showed no significant differences. Notably, the positivity rate of N antigen in peripheral blood peaked at 47.1% (95% CI: 37.8%-56.7%) during the first week of infection and then gradually decreased over time. Moreover, patients with severe COVID-19 exhibited significantly higher serum antigen levels than those with mild or moderate disease (p < 0.0001). Serum levels of SARS-CoV-2 S-RBD IgG and neutralizing antibodies (NAb) were also significantly higher in antigenemia-negative patients than in antigenemia-positive patients (p < 0.0001). CONCLUSIONS Our findings highlight the multifaceted role of antigenemia in SARS-CoV-2 and suggest its potential as a biomarker for disease monitoring and risk stratification.
Collapse
Affiliation(s)
- Jing Chen
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Shuai Zhao
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Haiyang Yan
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Yaomeng Huang
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Congzhen Wei
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Jiajia Liu
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China.
| | - Jingna Sun
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China.
| |
Collapse
|
9
|
Jiang Z, Pan M, Liu Y, Lundh T, Pineda D, Schenk L, Saber AT, Vogel U, Ljunggren S, Ricklund N, Engfeldt M, Krais AM, Broberg K. Integrative analyses of circulating microRNA expression profile in hexavalent chromium exposed workers - A cross-sectional study within the SafeChrom project. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137367. [PMID: 40098212 DOI: 10.1016/j.jhazmat.2025.137367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Exposure to hexavalent chromium (Cr(VI)) can occur during occupational activities and leading lung cancer. MicroRNA (miRNA) plays an important part in carcinogenesis. Whether Cr(VI) exposure causes cancer-related miRNA changes is yet uncharacterized. METHODS This study included 89 Cr(VI) exposed workers and 47 controls. MiRNAs were extracted from plasma followed by library preparations, miRNA sequencing, and differentially expressed miRNAs (DEMs) analysis. To understand the underlying biological functions, we used bioinformatics approaches, and qPCR was performed to validate the expression of potential target genes. RESULTS A total of 2100 miRNAs were detected. In the exposed workers, 59 DEMs were identified: 21 up-regulated and 38 down-regulated. Target genes for both up- and down-regulated DEMs were significantly enriched in: miRNAs in cancer, small cell lung cancer and non-small cell lung cancer. Protein-protein interactions showed a high number of interactions, in which CCNE2, CDK4 and E2F1 were predicted as hub genes, and the messenger RNA expression of those genes was significantly higher in the exposed workers compared with controls. CONCLUSIONS Our study suggests that low-to-moderate Cr(VI) exposure results in differential expression of lung-cancer-related miRNAs and associated target genes. Further studies are needed to validate our findings and clarify whether these changes predict cancer risk.
Collapse
Affiliation(s)
- Zheshun Jiang
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Mengyu Pan
- Cardiovascular Research-Translational Studies, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Yishan Liu
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Thomas Lundh
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Daniela Pineda
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Linda Schenk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anne T Saber
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Stefan Ljunggren
- Occupational and Environmental Medicine Center in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University
| | - Niklas Ricklund
- Department of Occupational and Environmental Medicine, Örebro University Hospital, Region Örebro County, Sweden
| | - Malin Engfeldt
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Department of Occupational and Environmental Medicine, Region Skåne, Lund, Sweden
| | - Annette M Krais
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Karin Broberg
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; National Research Centre for the Working Environment, Copenhagen, Denmark.
| |
Collapse
|
10
|
Cai Q, Jing C, Wang X, Xing X, Liu W. STEAP Proteins: Roles in disease biology and potential for therapeutic intervention. Int J Biol Macromol 2025; 309:142797. [PMID: 40185436 DOI: 10.1016/j.ijbiomac.2025.142797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Iron and copper are essential metal ions, and maintaining their metabolic balance is critical for organismal health. The Six-Transmembrane Epithelial Antigen of the Prostate (STEAP) protein family, comprising STEAP1, STEAP2, STEAP3, and STEAP4, plays a vital role in cellular metal homeostasis. These proteins are located on the cell membrane and are characterized by six transmembrane domains. With the exception of STEAP1, the STEAP proteins function as metal oxidoreductases due to their F420H2:NADP+ oxidoreductase (FNO)-like domain. However, STEAP1 contributes to metal metabolism through its heme group and interaction with other STEAP proteins. Beyond metal metabolism, STEAP proteins are involved in critical cellular processes, including the regulation of the cell cycle, proliferation, differentiation, and apoptosis. Notably, STEAP proteins are recognized as potential biomarkers and therapeutic targets in human cancers, particularly prostate cancer. This review outlines the structural features and functional roles of STEAP proteins in various diseases, including cancers, insulin resistance, non-alcoholic fatty liver disease (NAFLD), and benign prostatic hyperplasia, with a focus on their potential for therapeutic intervention.
Collapse
Affiliation(s)
- Qiaomei Cai
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, PR China
| | - Chao Jing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, PR China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin 300060, PR China
| | - Xiangling Xing
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China.
| | - Wancheng Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, PR China.
| |
Collapse
|
11
|
Chen Z, Zhong M, Lin Y, Zhang W, Zhu Y, Chen L, Huang Z, Luo K, Lu Z, Huang Z, Yan Y. METTL7B-induced histone lactylation prevents heart failure by ameliorating cardiac remodelling. J Mol Cell Cardiol 2025; 202:64-80. [PMID: 40068772 DOI: 10.1016/j.yjmcc.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
INTRODUCTION Lactylation is important for a variety of biological activities. It is reported that Class I histone deacetylases (HDAC1-3) are histone lysine delactylases. However, the role of lactylation in cardiac remodelling remains uncertain. OBJECTIVES To explore a novel regulator of lactylation and elucidate their functional mechanisms in cardiac remodelling and heart failure. METHODS GSE36961, GSE141910 and GSE174691 related to HCM (hypertrophic cardiomyopathy) were separately acquired from Gene expression Omnibus. Candidate genes related to both HCM and histone lactylation were determined by the intersection of DEGs (differentially expressed genes) and module genes sifted by WGCNA (Weighted Gene Co-Expression Network Analysis). METTL7B was screened out and its expression in hypertrophic myocardium was measured by qRT-PCR and western blotting. Furthermore, immunofluorescence, immunoprecipitation, and RNA pull-down assays were utilized to identify the biological functions of METTL7B. The myocardial biopsy of HCM and transverse aortic constriction (TAC) mouse model were performed to analyze the effects of METTL7B on cardiac remodelling in vivo. RESULTS We observed that the expression of METTL7B was down-regulated in hypertrophic myocardium, and the lactylation level was increased during the early stage and falling rapidly in the process of cardiac remodelling. Furthermore, we demonstrated that sodium lactate (NALA) administration fulfil a protective role on cardiac remodelling, and METTL7B alleviates cardiac remodelling and improves heart function by maintaining the activation of histone lactylation possibly at the later stage. Impressively, METTL7B suppressed the expression of USP38 via m6A dependent mRNA degradation, resulting in increasing ubiquitylation of HDAC3, which is a proven histone lysine delactylases. CONCLUSION We identifed METTL7B as a potential therapeutic target for myocardial remodelling and showed that it played a critical role in the promotion of myocardial lactylation, which is beneficial for improvement of cardiac function and attenuation of cardiac remodelling.
Collapse
Affiliation(s)
- Ziqi Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Meijun Zhong
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yuhui Lin
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Wei Zhang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yinghong Zhu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Lin Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Ziyao Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Kaiyuan Luo
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Zhifeng Lu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Zhaoqi Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Yi Yan
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
12
|
Voena C, Ambrogio C, Iannelli F, Chiarle R. ALK in cancer: from function to therapeutic targeting. Nat Rev Cancer 2025; 25:359-378. [PMID: 40055571 DOI: 10.1038/s41568-025-00797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 05/01/2025]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that acts as an oncogenic driver in solid and haematological malignancies in both children and adults. Although ALK-expressing (ALK+) tumours show strong initial responses to the series of ALK inhibitors currently available, many patients will develop resistance. In this Review, we discuss recent advances in ALK oncogenic signalling, together with existing and promising new modalities to treat ALK-driven tumours, including currently approved ALK-directed therapies, namely tyrosine kinase inhibitors, and novel approaches such as ALK-specific immune therapies. Although ALK inhibitors have changed the management and clinical history of ALK+ tumours, they are still insufficient to cure most of the patients. Therefore, more effort is needed to further improve outcomes and prevent the tumour resistance, recurrence and metastatic spread that many patients with ALK+ tumours experience. Here, we outline how a multipronged approach directed against ALK and other essential pathways that sustain the persistence of ALK+ tumours, together with potent or specific immunotherapies, could achieve this goal. We envision that the lessons learned from treating ALK+ tumours in the clinic could ultimately accelerate the implementation of innovative combination therapies to treat tumours driven by other tyrosine kinases or oncogenes with similar properties.
Collapse
Affiliation(s)
- Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fabio Iannelli
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy.
- Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Jiang F, Yang X, Shan L, Miao H, Shi C. 15-Deoxy-Δ-12,14-Prostaglandin J2 Represses Immune Escape of Lung Adenocarcinoma by Polarizing Macrophages Through Epidermal Growth Factor Receptor/Ras/Raf Pathway. J Immunother 2025; 48:119-126. [PMID: 39711151 DOI: 10.1097/cji.0000000000000546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
Lung adenocarcinoma (LUAD) is a widespread and deadly form of cancer. Prostaglandin 15-deoxy-Δ-12,14-prostaglandin J2 (15d-PGJ2) possesses antioxidant, anti-inflammatory, and anticancer properties. However, it is unclear whether this effect on LUAD progression stems from its ability to influence macrophage polarization. By utilizing 3- (4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry, colony formation, transwell assays, and enzyme linked immunosorbent assay (ELISA), we investigated how 15d-PGJ2 affects A549 cell viability, proliferation, apoptosis, and invasion, as well as levels of interleukin (IL)-4, IL-13, and IL-17. Human monocytic cell line THP-1 was induced into M2 macrophages using phorbol 12-myristate 13-acetate and IL-4/IL-13, followed by treatment with 15d-PGJ2. The study employed flow cytometry to observe the polarization of macrophages, quantitative reverse transcription polymerase chain reaction (qRT-PCR) to identify epidermal growth factor receptor (EGFR) expression, western blot for identifying expression of macrophage marker proteins, and examining EGFR/rat sarcoma (Ras)/rapidly accelerated fibrosarcoma (Raf) activation. In a coculture setup, CD8 + T cells were shown to have a proliferation capacity by carboxifluorescein diacetate succinimidyl ester (CFSE), a killing ability by lactate dehydrogenase, and an analysis of their interferon gamma and tumor necrosis factor alpha levels by ELISA. 15d-PGJ2 reduced invasion capacity and expression of inflammatory cytokines, lowered A549 cell viability in a dose-dependent way, and promoted apoptosis. 15d-PGJ2 facilitated the transition of M2 macrophages to the M1 type, inhibited Ras/Raf pathway activation, reduced EGFR expression in macrophages, and stimulated CD8 + T cells to enhance anti-tumor immunity. 15d-PGJ2 repressed M2 macrophage polarization and LUAD immune evasion by targeting the EGFR/Ras/Raf pathway in macrophages.
Collapse
Affiliation(s)
- Fan Jiang
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| | - Xiaoxiao Yang
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| | - Liqun Shan
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| | - Huiwen Miao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaohong Shi
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
14
|
Atçeken Z, Çelik Y, Atasoy Ç, Peker Y. Artificial Intelligence-guided Total Opacity Scores and Obstructive Sleep Apnea in Adults with COVID-19 Pneumonia. THORACIC RESEARCH AND PRACTICE 2025; 26:107-114. [PMID: 39930690 PMCID: PMC12047196 DOI: 10.4274/thoracrespract.2024.24090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/30/2024] [Indexed: 05/03/2025]
Abstract
OBJECTIVE We previously demonstrated that artificial intelligence (AI)-directed chest computed tomography (CT)-based total opacity scores (TOS) are associated with high-risk obstructive sleep apnea (OSA) based on the Berlin Questionnaire. In the current study, we examined the association between TOS severity and OSA severity based on polysomnography (PSG) recordings among participants with a history of Coronavirus disease-2019 (COVID-19) infection. MATERIAL AND METHODS This was a post-hoc analysis of 56 patients who underwent CT imaging after being diagnosed with COVID-19 pneumonia as well as overnight PSG for a validation study with a median of 406 days after the initial COVID-19 onset. The AI software quantified the overall opacity scores, which included consolidation and ground-glass opacity regions on CT scans. TOS was defined as the volume of high-opacity regions divided by the volume of the entire lung, and severe TOS was defined as the score ≥15. OSA was defined as an apnea-hypopnea index (AHI) of at least 15 events/h. RESULTS In total, 21 participants had OSA and 35 had no OSA. The median TOS was 10.5 [interquartile range (IQR) 1.6-21.2] in the OSA group and 2.8 (IQR 1.4-9.0) in the non-OSA group (P = 0.047). In a multivariate logistic regression analysis, OSA, AHI, and oxygen desaturation index were associated with severe TOS (P < 0.05 for all, respectively) adjusted for age, sex, body mass index, and hypertension. CONCLUSION AI-directed CT-based TOS severity in patients with COVID-19 pneumonia was associated with OSA severity based on PSG recordings. These results support our previous findings suggesting an association between questionnaire-based high-risk OSA and worse outcomes in COVID-19 pneumonia.
Collapse
Affiliation(s)
- Zeynep Atçeken
- Department of Radiology, Koç University Faculty of Medicine, İstanbul, Türkiye
| | - Yeliz Çelik
- Department of Pulmonary Medicine, Koç University Faculty of Medicine; Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
| | - Çetin Atasoy
- Department of Radiology, Koç University Faculty of Medicine, İstanbul, Türkiye
| | - Yüksel Peker
- Department of Pulmonary Medicine, Koç University Faculty of Medicine; Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Sciences, Respiratory Medicine and Allergology, Lund University Faculty of Medicine, Lund, Sweden
- Department of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pennsylvania, USA
| |
Collapse
|
15
|
Jaiswal A, Fervers P, Meng F, Zhang H, Móré D, Giannakis A, Wailzer J, Bucher AM, Maintz D, Kottlors J, Shahzad R, Persigehl T. Performance of AI Approaches for COVID-19 Diagnosis Using Chest CT Scans: The Impact of Architecture and Dataset. ROFO-FORTSCHR RONTG 2025. [PMID: 40300640 DOI: 10.1055/a-2577-3928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
AI is emerging as a promising tool for diagnosing COVID-19 based on chest CT scans. The aim of this study was the comparison of AI models for COVID-19 diagnosis. Therefore, we: (1) trained three distinct AI models for classifying COVID-19 and non-COVID-19 pneumonia (nCP) using a large, clinically relevant CT dataset, (2) evaluated the models' performance using an independent test set, and (3) compared the models both algorithmically and experimentally.In this multicenter multi-vendor study, we collected n=1591 chest CT scans of COVID-19 (n=762) and nCP (n=829) patients from China and Germany. In Germany, the data was collected from three RACOON sites. We trained and validated three COVID-19 AI models with different architectures: COVNet based on 2D-CNN, DeCoVnet based on 3D-CNN, and AD3D-MIL based on 3D-CNN with attention module. 991 CT scans were used for training the AI models using 5-fold cross-validation. 600 CT scans from 6 different centers were used for independent testing. The models' performance was evaluated using accuracy (Acc), sensitivity (Se), and specificity (Sp).The average validation accuracy of the COVNet, DeCoVnet, and AD3D-MIL models over the 5 folds was 80.9%, 82.0%, and 84.3%, respectively. On the independent test set with n=600 CT scans, COVNet yielded Acc=76.6%, Se=67.8%, Sp=85.7%; DeCoVnet provided Acc=75.1%, Se=61.2%, Sp=89.7%; and AD3D-MIL achieved Acc=73.9%, Se=57.7%, Sp=90.8%.The classification performance of the evaluated AI models is highly dependent on the training data rather than the architecture itself. Our results demonstrate a high specificity and moderate sensitivity. The AI classification models should not be used unsupervised but could potentially assist radiologists in COVID-19 and nCP identification. · This study compares AI approaches for diagnosing COVID-19 in chest CT scans, which is essential for further optimizing the delivery of healthcare and for pandemic preparedness.. · Our experiments using a multicenter, multi-vendor, diverse dataset show that the training data is the key factor in determining the diagnostic performance.. · The AI models should not be used unsupervised but as a tool to assist radiologists.. · Jaiswal A, Fervers P, Meng F et al. Performance of AI Approaches for COVID-19 Diagnosis Using Chest CT Scans: The Impact of Architecture and Dataset. Rofo 2025; DOI 10.1055/a-2577-3928.
Collapse
Affiliation(s)
- Astha Jaiswal
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Fervers
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fanyang Meng
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
| | - Huimao Zhang
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
| | - Dorottya Móré
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Athanasios Giannakis
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Jasmin Wailzer
- Institute for Diagnostic and Interventional Radiology, Frankfurt University Hospital, Frankfurt, Germany
| | - Andreas Michael Bucher
- Institute for Diagnostic and Interventional Radiology, Frankfurt University Hospital, Frankfurt, Germany
| | - David Maintz
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jonathan Kottlors
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Rahil Shahzad
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Philips Healthcare, Innovative Technologies, Aachen, Germany
| | - Thorsten Persigehl
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
16
|
Liu XF, Song B, Sun CB, Zhu Q, Yue JH, Liang YJ, He J, Zeng XL, Qin YC, Chen QY, Mai HQ, Zhang X, Li J. Tumor-infiltrated double-negative regulatory T cells predict outcome of T cell-based immunotherapy in nasopharyngeal carcinoma. Cell Rep Med 2025:102096. [PMID: 40315843 DOI: 10.1016/j.xcrm.2025.102096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/24/2025] [Accepted: 04/04/2025] [Indexed: 05/04/2025]
Abstract
Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) has demonstrated clinical success in solid tumors. We analyze 47 TIL infusion products and 62 pretreatment tumor microenvironments (TMEs) from a randomized phase 2 clinical study of concurrent chemoradiotherapy plus TIL-ACT (NCT02421640). Using single-cell and bulk RNA sequencing along with flow cytometry, we identify 14 CD3+ T cell clusters within 26 TIL infusion products: 11 CD3+CD8+ TILs, 2 CD3+CD4+ TILs, and 1 CD3+CD8-CD4- double-negative (DN) TIL. (DN) TILs, significantly associated with poor TIL-ACT outcomes, exhibit an activated regulatory T cell-like phenotype and include two CD56+ and four CD56- subsets. Among them, CD56-KZF2+ (DN) TILs are predominantly suppressive. (DN) TILs inhibit CD8+ TIL expansion via Fas-FasL, transforming growth factor β (TGF-β), and interleukin (IL)-10 signaling. Distinct CD8+ T subsets differentially impact on TIL-ACT outcomes, while 9 baseline TME gene signatures and 14 intracellular T cell genes hold prognostic value. Our findings identify predictive TIL subsets and biomarkers for TIL-ACT outcomes.
Collapse
Affiliation(s)
- Xiu-Feng Liu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Bin Song
- BGI, Shenzhen 518083, P.R. China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Chang-Bin Sun
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, P.R. China
| | - Qian Zhu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | | | - Yu-Jing Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jia He
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Xi-Liang Zeng
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | | | - Qiu-Yan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Xi Zhang
- BGI, Shenzhen 518083, P.R. China.
| | - Jiang Li
- Department of Biotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
17
|
Wei H, Wang S, Wang S, Xiao W, Ding A, Deng N, Wang H. Efficient detection of SARS-CoV-2 IgG by a smartphone-based high-throughput fiber optic sensor. Talanta 2025; 294:128183. [PMID: 40305926 DOI: 10.1016/j.talanta.2025.128183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
The World Health Organization has indicated that Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2) is likely to persist for an extended period, with the potential emergence of new mutated strains. To effectively control the spread of the virus, there is an urgent need for simple, rapid, and reliable assays to identify SARS-CoV-2 infections, particularly in community hospitals or clinical centers. This study introduces a smartphone-based high-throughput fiber-optic immunosensor (HFIS) for rapid SARS-CoV-2 IgG detection. The system significantly improves upon existing methods by combining laboratory-level accuracy with point-of-care convenience. Key advantages include: (1) 100-fold higher throughput than typical smartphone lateral flow readers (100 vs 1 sample/run); (2) 3 × faster than smartphone-ELISA hybrids (40 vs 120+ minutes); (3) Enhanced sensitivity with 86.05 % detection rate versus 83.72 % for commercial ELISA; (4) Lower cost than multi-well smartphone readers by eliminating movable parts; (5) True portability as a 259g standalone device versus bulkier smartphone attachments; and (6) Laboratory-level accuracy (AUC = 0.96) surpassing most smartphone-based assays (typically AUC<0.90).This integrated solution addresses critical needs for large-scale screening, offering rapid, high-throughput testing with laboratory-quality results in field settings. The technology's modular design also allows potential adaptation for other infectious disease diagnostics.
Collapse
Affiliation(s)
- Hongyu Wei
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Bioengineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Shangwu Wang
- Department of Clinical Laboratory, Taojiang People's Hospital, Yiyang, 413400, China
| | - Si Wang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Bioengineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei Xiao
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Aihua Ding
- Department of Clinical Laboratory, Taojiang People's Hospital, Yiyang, 413400, China.
| | - Ning Deng
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Bioengineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong Wang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Bioengineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
18
|
Wu B, Zhu J, Chen L, Wang X, Zhang H, Guan K, Li Y. The Relationship Between Preoperative Neutrophil-Lymphocyte Ratio and Postoperative Length of Stay in Carotid Body Tumor Resection. Int J Genomics 2025; 2025:5431545. [PMID: 40314030 PMCID: PMC12043386 DOI: 10.1155/ijog/5431545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Carotid body tumor (CBT) resection is a complex surgical procedure often resulting in extended postoperative length of stay (PLOS) due to potential nerve injuries, arterial damage, and wound complications. The neutrophil-to-lymphocyte ratio (NLR) is a known marker of systemic inflammation and has been associated with adverse outcomes in various surgical settings. However, the relationship between preoperative NLR and PLOS in CBT patients has not been explored. This study aims to investigate the association between preoperative NLR and PLOS in CBT resections, particularly examining whether elevated NLR correlates with longer hospital stays and potentially hinders recovery. In this retrospective cohort study, we analyzed data from 231 CBT patients who underwent resection at Changhai Hospital, Shanghai, between 2008 and 2020. Patients were grouped based on their PLOS (short, medium, and long stays), and NLR was calculated from peripheral blood samples taken preoperatively. Univariate and multivariate regression models adjusted for sociodemographic and operative factors, including Shamblin classification, were used to examine the relationship between NLR and PLOS. Elevated preoperative NLR has been found to be significantly correlated with prolonged PLOS, with each incremental increase in NLR corresponding to an approximate extension of 0.12 days in PLOS after adjusting for confounding factors. Stratified analysis revealed that this association was most pronounced in patients with Shamblin II tumors, likely due to the moderate tumor size and adhesion in these cases, which necessitates more extensive dissection and increases vulnerability to nerve injury. Elevated preoperative NLR may serve as a predictor of prolonged recovery in CBT resections, particularly for Shamblin II cases. This finding highlights the potential utility of NLR in preoperative assessment and patient management to optimize surgical timing and reduce hospital stays. Further research with larger cohorts is needed to confirm the predictive value of NLR and explore its clinical application in surgical planning for CBT patients.
Collapse
Affiliation(s)
- Biao Wu
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China
| | - Jiang Zhu
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China
| | - Liang Chen
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China
| | - Xiaonan Wang
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China
| | - Hao Zhang
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China
| | - Kunyu Guan
- Pediatrics, Changhai Hospital, Shanghai, China
| | - Yu Li
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China
| |
Collapse
|
19
|
Ma Q, Zhu Y, Zhang D, Su X, Jiang C, Zhang Y, Zhang X, Han N, Shu G, Yin G, Wang M. Reprogramming and targeting of cholesterol metabolism in tumor-associated macrophages. J Mater Chem B 2025. [PMID: 40266660 DOI: 10.1039/d5tb00236b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Cholesterol, as a major component of cell membranes, is closely related to the metabolic regulation of cells and organisms; tumor-associated macrophages play an important push role in tumor progression. We know that tumor-associated macrophages are polarized from macrophages, and the abnormalities of cholesterol metabolism that may be induced during their polarization are worth discussing. This manuscript focuses on metabolic abnormalities in tumor-associated macrophages, and first provides a basic summary of the regulatory mechanisms of abnormal macrophage polarization. Subsequently, it comprehensively describes the features of abnormal glucose, lipid and cholesterol metabolism in TAMs as well as the different regulatory pathways. Then, the paper also discusses the link between abnormal cholesterol metabolism in TAMs and tumors, chronic diseases and aging. Finally, the paper summarizes cancer therapeutic strategies targeting cholesterol metabolism that are already in clinical trials, as well as nanomaterials capable of targeting cholesterol metabolism that are in the research stage, in the hope of providing value for the design of targeting materials. Overall, elucidating metabolic abnormalities in tumor-associated macrophages, particularly cholesterol metabolism, could provide assistance in tumor therapy and the design of targeted drugs.
Collapse
Affiliation(s)
- Qiaoluo Ma
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Ying Zhu
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Dongya Zhang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Xiaohan Su
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Can Jiang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Yuzhu Zhang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Xingting Zhang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Na Han
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Guang Shu
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| |
Collapse
|
20
|
Campodónico VL, Ruelle J, Fitzgerald A, Bergman Y, Osborne B, Bourdas D, Lu J, Carroll KC, Simner PJ. Evaluation of long-read 16S rRNA next-generation sequencing for identification of bacterial isolates in a clinical diagnostic laboratory. J Clin Microbiol 2025:e0167024. [PMID: 40261041 DOI: 10.1128/jcm.01670-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
Sanger sequencing of the first ~500 bp of the 16S rRNA gene is frequently used to identify bacterial pathogens that have ambiguous biochemical profiles or proteomic mass spectra. When diversity does not occur within that region, genus-level and/or species-level identification may not be possible, and a longer sequence or alternative target may be required to distinguish between genera/species. In this study, we evaluated a clinically relevant end-to-end solution for long-read (~1,500 nt) 16S rRNA next-generation sequencing by Oxford Nanopore Technologies (ONT) compared to a ~500 nt Sanger sequencing approach for the identification of 153 bacterial clinical isolates. Sequencing data were analyzed using the IDNS software from SmartGene and its proprietary 16S Centroid reference database (Centroid database) SmartGene software and the Centroid database. The agreement of the two platforms on species- and genus-level identification was determined, and discrepancies were resolved by whole-genome sequencing. ONT had a higher taxonomic resolution at the genus level (P < 0.01). When genus-level identification was achieved by both methods, concordance to the best matching genus was 100%. When species-level identification was achieved by both methods, concordance to the best matching species was 91%. The costs per test were ~$25.30 (when multiplexing 24 samples/run) and $74 for ONT and Sanger sequencing, respectively. The hands-on time spent performing sequencing was similar for both methods, but the turnaround time of ONT was significantly shorter than that of Sanger sequencing.IMPORTANCEThis study adds to existing literature by describing a validated end-to-end solution of 16S rRNA gene Oxford Nanopore sequencing for bacterial isolate identification, including sequencing run time evaluation, automated analysis (SmartGene 16S Identification App) and interpretation of results, that can be incorporated into clinical and public health laboratories with a simple and cost-effective workflow.
Collapse
Affiliation(s)
- Victoria L Campodónico
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jean Ruelle
- SmartGene Services, EPFL Innovation Park, Lausanne, Switzerland
| | - Anna Fitzgerald
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yehudit Bergman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brenda Osborne
- SmartGene Services, EPFL Innovation Park, Lausanne, Switzerland
| | - Dimitrios Bourdas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Lu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karen C Carroll
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Patricia J Simner
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Tran ET, Patel RA, Chariyamane A, Ray RB. Long non-coding RNAs as therapeutic targets in head and neck squamous cell carcinoma and clinical application. FEBS Open Bio 2025. [PMID: 40231344 DOI: 10.1002/2211-5463.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a major global health burden, often associated with poor prognosis and limited therapeutic options. Long non-coding RNAs (lncRNAs), a diverse group of non-coding RNA molecules > 200 nucleotides in length, have emerged as critical regulators in the pathogenesis of HNSCC. This review summarizes the mechanisms through which certain lncRNAs regulate chromatin modification, mRNA splicing, and interactions with RNA-binding proteins and contribute to the development and progression of HNSCC. Interaction of lncRNAs with key oncogenic pathways, such as PI3K/AKT and Wnt/β-catenin, highlights their importance in tumor progression. The role of lncRNAs, such as ELDR, MALAT1, NEAT1, HOTAIR, and UCA1, which promote cell proliferation, metastasis, immune evasion, and therapy resistance is discussed. Moreover, several lncRNAs are being evaluated in clinical trials for their potential as biomarkers, reflecting their clinical significance. We further address the challenges and opportunities for targeting lncRNA therapeutically, highlighting the promise of lncRNA-based interventions for personalized cancer treatment. Gaining insight into the function of lncRNAs in HNSCC could pave the way for novel therapeutic strategies to potentially improve patient outcomes.
Collapse
Affiliation(s)
- Ellen T Tran
- Department of Pathology, Saint Louis University, MO, USA
| | - Ruchi A Patel
- Department of Pathology, Saint Louis University, MO, USA
| | | | - Ratna B Ray
- Department of Pathology, Saint Louis University, MO, USA
| |
Collapse
|
22
|
Ye W, Zhang X, Tang Z, Hu Y, Zheng Y, Yuan Y. Comprehensive analysis of glycometabolism-related genes reveals PLOD2 as a prognostic biomarker and therapeutic target in gastric cancer. BMC Gastroenterol 2025; 25:256. [PMID: 40229676 PMCID: PMC11998276 DOI: 10.1186/s12876-025-03878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide, with limited therapeutic options and a poor prognosis, particularly in advanced stages. Glycometabolism, a hallmark of cancer, plays a critical role in tumor progression, immune evasion, and response to therapy. However, the specific roles of glycometabolism-related genes and their prognostic and therapeutic implications in GC remain inadequately understood. METHODS Transcriptomic and clinical data from GC patients were retrieved from TCGA and GEO databases. Glycometabolism-related genes were identified and analyzed using machine learning algorithms to construct a prognostic model. Functional assays, immune profiling, and pathway enrichment analyses were performed to explore the roles of these genes in tumor progression, immune-modulatory effects, and drug resistance. PLOD2, the gene with the highest prognostic significance, was further investigated to uncover its underlying regulatory mechanisms, roles in immune modulation, and contribution to therapeutic resistance. RESULTS A glycometabolism-related prognostic model consisting of four genes (PLOD2, CHSY3, SLC2A3 and SLC5A1) was developed and validated, effectively stratifying GC patients into high- and low-risk subgroups with distinct survival outcomes. Among these, PLOD2 emerged as the most significant gene, exhibiting strong associations with tumor progression and poor survival. Functional analyses revealed that PLOD2 promotes glycolysis and tumor progression through activation of the PI3K/AKT/mTOR pathway. Immune profiling revealed that PLOD2 overexpression is associated with an immunosuppressive tumor microenvironment, characterized by increased M2 macrophage infiltration and reduced immune activity. Moreover, treatment with rapamycin, an mTOR inhibitor, significantly suppressed PLOD2-mediated proliferation and anchorage-independent growth in GC cells, highlighting the central role of the PI3K/AKT/mTOR pathway in PLOD2-driven oncogenic behaviors. CONCLUSIONS This study identifies PLOD2 as a key prognostic biomarker and therapeutic target in gastric cancer. As a central component in a glycometabolism-related model, PLOD2 promotes glycolysis, tumor progression, and immune evasion via the PI3K/AKT/mTOR pathway. The model effectively stratifies patient risk, offering both prognostic utility and therapeutic insight. Targeting PLOD2-mediated pathways may represent a promising strategy for precision therapy and improved clinical outcomes in gastric cancer.
Collapse
Affiliation(s)
- Wanchun Ye
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Xiaolei Zhang
- Department of Clinical Laboratory, Jinan Fourth People's Hospital, Jinan, China
| | - Zhongjie Tang
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yufeng Hu
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yuanliang Zheng
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yuping Yuan
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China.
| |
Collapse
|
23
|
Orlandella FM, Arcone R, Luciano N, Salvatore G, Motti ML. Novel Biological Strategies for Melanoma Therapy: A Focus on lncRNAs and Their Targeting. Cancers (Basel) 2025; 17:1273. [PMID: 40282449 PMCID: PMC12025846 DOI: 10.3390/cancers17081273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Increasing evidence revealed that restoring the correct expression of lncRNAs could have implications in the management of melanoma patients. In this context, here, we aim to dissect the main characteristics of lncRNAs altered in melanoma and their crosstalk with the signaling pathways involved in the progression of this disease. We also highlight the role of nucleic acid-based techniques and natural compounds (i.e., phytochemicals) as a therapeutic tool to increase or silence their expression in cancer cells. Finally, we explore the advances in nanotechnologies as delivery systems to efficiently carry these chemicals into cancer cells, thus limiting their potential off-target effects. The analysis of the literature showed that HOTAIR, MALAT1, and H19 are the oncogenic lncRNAs most studied in melanoma, while MEG3 is an important tumor suppressor decreased in this cancer. The aberrant expression of these lncRNAs affects several hallmarks of cancer, e.g., proliferation, motility, and epithelial to mesenchymal transition, promoting the melanoma plasticity and drug resistance. In this frame, siRNA, antisense oligonucleotide, and CRISPR-Cas9 genome editing appear to be the most effective nucleic acid strategies to restore the physiologic expression of lncRNA, while curcumin, resveratrol, and quercetin are the main phytochemicals able to target and influence the expression of lncRNAs altered in cancer. Overall, this study provides a comprehensive overview regarding the role of lncRNAs in the phenotype plasticity of melanoma cells and their potential targeting using RNA-based therapy and natural products.
Collapse
Affiliation(s)
- Francesca Maria Orlandella
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (F.M.O.); (R.A.); (N.L.)
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, 80131 Naples, Italy
| | - Rosaria Arcone
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (F.M.O.); (R.A.); (N.L.)
| | - Neila Luciano
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (F.M.O.); (R.A.); (N.L.)
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, 80131 Naples, Italy
| | - Giuliana Salvatore
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (F.M.O.); (R.A.); (N.L.)
- CEINGE-Biotecnologie Avanzate “Franco Salvatore”, 80131 Naples, Italy
| | - Maria Letizia Motti
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (F.M.O.); (R.A.); (N.L.)
| |
Collapse
|
24
|
Luo AL, Zheng WY, Zhang Q, Yuan Y, Li MQ, Du K, Gao AR, Pei LJ, Xie J, Chen WH, Zhang L, Guo XZ, Yang XR, Zeng C, Yang GH, Deng M. COPS5 Triggers Ferroptosis Defense by Stabilizing MK2 in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416360. [PMID: 40198582 DOI: 10.1002/advs.202416360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sorafenib, which is proven to serve as a potent ferroptosis inducer, is used as a first-line treatment for patients with advanced hepatocellular carcinoma (HCC), but it has limited clinical benefits, mainly due to drug resistance. Herein, using genome-wide CRISPR/Cas9 knockout screening and multiple functional studies, this work identifies COP9 signalosome subunit 5 (COPS5) as a driver of sorafenib resistance and a suppressor of ferroptosis in HCC. Consistently, the amplification and overexpression of COPS5 are frequently observed in clinical HCC samples, which are associated with poor patient prognosis and might predict patient response to sorafenib therapy. Mechanistically, COPS5 stabilized mitogen-activated protein kinase 2 (MK2) through deubiquitination and, in turn, induced the activation of heat shock protein beta-1 (HSPB1), a ferroptosis repressor, thereby protecting HCC cells from ferroptosis and consequently leading to sorafenib resistance and tumor progression, while its own expression could be induced by sorafenib treatment via activating transcription factor 4 (ATF4)-activated transcription. Furthermore, pharmacological inhibition of COPS5/MK2 synergize with sorafenib to induce ferroptosis and suppress HCC progression. This data reveals the crucial role of COPS5 in triggering ferroptosis defense and sorafenib resistance through the activation of the MK2-HSPB1 axis in HCC and highlights the potential of targeting COPS5/MK2 combined with sorafenib as a promising strategy for treating HCC.
Collapse
Affiliation(s)
- Ai-Ling Luo
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Wen-Ying Zheng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Qiong Zhang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Yan Yuan
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Mei-Qi Li
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Kai Du
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - An-Ran Gao
- Department of Laboratory Medicine, Shunde Hospital, Guangzhou University of Chinese Medicine, Foshan, 528300, China
| | - Li-Jun Pei
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Jie Xie
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Wen-Hao Chen
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Long Zhang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiu-Zhu Guo
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiao-Ran Yang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Chao Zeng
- Department of Pathology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Guo-Hua Yang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Min Deng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| |
Collapse
|
25
|
Sun A, Stejskalová P, Liu X, Řezníček T, Brodský J, Gablech I, Zítka O, Neužil P. Two colorimetric LAMP systems for nucleic acid-based diagnostics. Anal Chim Acta 2025; 1346:343752. [PMID: 40021320 DOI: 10.1016/j.aca.2025.343752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/22/2025] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND This study introduces an advanced 8-well loop-mediated isothermal amplification (LAMP) system specifically designed for the automated colorimetric detection of SARS-CoV-2. Incorporating two distinct configurations having either three light-emitting diodes (LEDs) with varying emission wavelengths per well, paired with a photodiode detector, or utilizing white LED illumination with a red, green, and blue (RGB) sensor. The colorimetric LAMP aims to provide a more accessible and rapid diagnostic tool than traditional fluorescence methods due to the system's simplicity. RESULTS We designed, assembled, and compared two colorimetric home-assembled LAMP systems, the first one based on three LEDs, each with a different color with a photodiode, and the second one having RGB and a white LED, with traditional fluorescence-based LAMP method performed on a commercial qPCR instrument. Results demonstrated that the colorimetric RT-LAMP assays achieved critical threshold time (CT), closely matching the CT value of fluorescence-based detection accomplished by the qPCR instrument. We performed the fundamental experiment employing an identical RNA copy number of 1,570copies·μL-1, getting the CT value of (16.70 ± 0.43) min (mean ± standard deviation from 23 measurements). Then, we also performed different RNA numbers of copies between the highest and lowest RNA contents of ≈ 157,000 copies·μL-1 and ≈ 1570 copies·μL-1, respectively, getting CT values from (13.30 ± 0.04) min to (13.75 ± 0.30) min and (17.04 ± 0.02) min to (17.26 ± 0.02) min, all (mean ± standard deviation from three measurements). The colorimetric systems demonstrated rapid response and precision across varied viral loads while keeping the system simple due to the colorimetric detection method. SIGNIFICANCE AND NOVELTY The LAMP system's rapid and precise detection capabilities underscore its potential as an effective tool for point-of-need diagnostics. It is crucial for timely responses in ongoing and future pandemic scenarios. This system enhances testing accessibility and provides a robust platform for potential adaptation to other pathogenic threats, making it a valuable asset in global health diagnostics.
Collapse
Affiliation(s)
- Antao Sun
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, School of Mechanical Engineering, Northwestern Polytechnical University, 127 West Youyi Road, 710072, Xi'an, Shaanxi, PR China
| | - Petra Stejskalová
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemědělská 1, 61300, Brno, Czech Republic
| | - Xiaocheng Liu
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, School of Mechanical Engineering, Northwestern Polytechnical University, 127 West Youyi Road, 710072, Xi'an, Shaanxi, PR China
| | - Tomáš Řezníček
- ITD Tech s.r.o, Osvoboditelu 1005, 735 81, Bohumín, Czech Republic
| | - Jan Brodský
- Department of Microelectronics, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technická 3058/10, Brno, 61600, Czech Republic
| | - Imrich Gablech
- Department of Microelectronics, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technická 3058/10, Brno, 61600, Czech Republic
| | - Ondřej Zítka
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemědělská 1, 61300, Brno, Czech Republic
| | - Pavel Neužil
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, School of Mechanical Engineering, Northwestern Polytechnical University, 127 West Youyi Road, 710072, Xi'an, Shaanxi, PR China.
| |
Collapse
|
26
|
Liu L, Li X, Yang H, Xu F, Dong X. Bioinformatic Analysis of Apoptosis-Related Genes in Preeclampsia Using Public Transcriptomic and Single-Cell RNA Sequencing Datasets. J Inflamm Res 2025; 18:4785-4812. [PMID: 40224388 PMCID: PMC11992479 DOI: 10.2147/jir.s507660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose Apoptosis, which is crucial in preeclampsia (PE), affects trophoblast survival and placental function. We used transcriptomics and single-cell RNA sequencing (scRNA-seq) to explore apoptosis-related genes (ARGs) and their cellular mechanisms as potential PE biomarkers. Patients and Methods All the data included in this study were sourced from public databases. We used scRNA-seq and differential expression analysis, combined with five algorithms from the CytoHubba plugin, to identify ARGs as PE biomarkers. These were integrated into diagnostic nomograms. Mechanistic studies involved enrichment analysis and immune profiling. Biomarker expression was examined at the single-cell level, and verified in clinical samples by RT-qPCR. Results Cluster of Differentiation 44 (CD44), Macrophage migration inhibitory factor (MIF), PIK3R1, and Toll-like receptor 4 (TLR4) were identified as PE biomarkers. CD44 and TLR4 were down-regulated, while MIF and PIK3R1 were up-regulated. When integrated into the diagnostic nomogram, they showed clinical utility and affected cell functions. In the immune profile of PE, monocytes decreased, resting NK cells increased, and the activities of APC, checkpoint, T-cell co-stimulation, and MHC class I pathways reduced. ScRNA-seq identified 11 cell types, 10 of which were significantly different. Endothelial cell communication with other cell types decreased, while the interaction between common myeloid progenitors (CMP) and villous cytotrophoblasts (VCT) enhanced. The expression levels of CD44, MIF, and PIK3R1 in VCT were significantly different and key to PE. Their decrease in early PE and increase in late PE reflected the placenta's adaptation to adverse pregnancy conditions. Conclusion Four ARGs, CD44, MIF, PIK3R1, and TLR4, identified through comprehensive analyses, served as significant biomarkers for PE and offered insights into PE's cellular mechanisms of PE, providing valuable references for further research.
Collapse
Affiliation(s)
- Lingyan Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Xiuling Li
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Hongfen Yang
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Fei Xu
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Department of Pain Management, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Xudong Dong
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| |
Collapse
|
27
|
Huang M, Zhang W, Dong J, Hu Z, Tan X, Li H, Sun K, Zhao A, Huang T. Genome-Wide Association Studies of Body Weight and Average Daily Gain in Chinese Dongliao Black Pigs. Int J Mol Sci 2025; 26:3453. [PMID: 40244387 PMCID: PMC11989284 DOI: 10.3390/ijms26073453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
In the domain of swine production, body weight (BW) and average daily gain (ADG) are recognized as the primary performance indicators. Nevertheless, the genetic architecture of ADG and BW in Dongliao black (DLB) pigs remains to be fully elucidated. In this study, we performed a genome-wide association analysis of BW, ADG, and body mass index (BMI) in 358 DLB pigs of different days of age. The genome-wide association study (GWAS) showed the following: (1) The most significant single nucleotide polymorphism (SNP) detected for BW was on Sus scrofa chromosome (SSC) 11:100,808 (p-value = 1.16 × 10-6) that was also the most significant SNP for ADG. (2) The most significant SNP associated with BMI was SSC17:51,463,521 (p-value = 5.16 × 10-8). (3) SNPs SSC10:6,523,844 and SSC17:23,852,682 were identified in both BW and ADG. A meta-analysis was conducted on BW at different days and demonstrated SSC5:39,028,335 (p-value = 8.37 × 10-6) which was not identified in the results of each single trait. The regions of two SNPs (SSC11:100,808, SSC4:10,703,277) exhibited considerable influence on both BW and ADG and the related regions were selected for linkage disequilibrium (LD) analyses that exhibited a notable linkage. In addition, several genes were identified that are associated with obesity and play roles in lipid metabolism, including MACROD2, PHLPP2, CYP2E1, and STT3B.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ayong Zhao
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China; (M.H.); (W.Z.); (J.D.); (Z.H.); (X.T.); (H.L.); (K.S.)
| | - Tao Huang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China; (M.H.); (W.Z.); (J.D.); (Z.H.); (X.T.); (H.L.); (K.S.)
| |
Collapse
|
28
|
Bai X, Liu R, Tang Y, Yang L, Niu Z, Hu Y, Zhang L, Chen M. Combined Transcriptomic and Mendelian Randomisation Explores the Diagnostic Value of Ubiquitination-Related Genes in Sepsis. J Inflamm Res 2025; 18:4709-4724. [PMID: 40201575 PMCID: PMC11977632 DOI: 10.2147/jir.s489077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Sepsis is the 10th leading cause of death globally and the most common cause of death in patients with infections. Ubiquitination plays a key role in regulating immune responses during sepsis. This study combined bioinformatics and Mendelian randomization (MR) analyses to identify ubiquitin-related genes (UbRGs) with unique roles in sepsis. Methods Relevant genes were obtained from the GSE28750 dataset and GSE95233, weighted gene co-expression network analyses were performed to identify gene modules, and differentially expressed UBRGs (DE-UBRGs) were generated by differentially expressed genes (DEGs) crossover with key modular genes and UBRGs in sepsis and normal samples. Causal relationships between sepsis and UbRGs were analysed using MR, performance diagnostics were performed using subject work characteristics (ROC) curves, and an artificial neural network (ANN) model was developed. On this basis, immune infiltration was performed and the expression of key genes was verified in animal models. Results 3022 DEGs were found between sepsis and normal. A total of 2620 genes were obtained as key modular genes. Crossing DEGs, key modular genes and UBRGs yielded 93 DE-UBRGs. MR results showed WDR26 as a risk factor for sepsis (OR>1) and UBE2D1 as a protective factor for sepsis (OR<1), which was reinforced by scatterplot and forest plot. ROC curves showed that WDR26 and UBE2D1 could accurately differentiate between sepsis and normal samples. Confusion matrix and ROC curve results indicate that the artificial neural network model has strong diagnostic ability. The results of immune infiltration showed that.WDR26 was negatively correlated with plasma cells, while UBE2D1 was positively correlated with CD4 naïve T cells. Significant differences between sepsis and normal were obtained between UBE2D1 and WDR26 in the animal model. Conclusion There appeared to be a causal relationship between sepsis, WDR26 and UBE2D1. The insights were of value for effective clinical diagnosis and treatment in sepsis.
Collapse
Affiliation(s)
- Xue Bai
- Department of Emergency, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, People’s Republic of China
| | - RuXing Liu
- Department of Emergency, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Yujiao Tang
- Department of Emergency, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - LiTing Yang
- Department of Emergency, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, People’s Republic of China
| | - Zesu Niu
- Department of Emergency, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Yi Hu
- Department of Emergency, The Third Clinical Medical College of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Ling Zhang
- Department of Emergency, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, People’s Republic of China
| | - MengFei Chen
- Department of Emergency, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, People’s Republic of China
| |
Collapse
|
29
|
Carandang THDC, Cunanan DJ, Co GS, Pilapil JD, Garcia JI, Restrepo BI, Yotebieng M, Torrelles JB, Notarte KI. Diagnostic accuracy of nanopore sequencing for detecting Mycobacterium tuberculosis and drug-resistant strains: a systematic review and meta-analysis. Sci Rep 2025; 15:11626. [PMID: 40185766 PMCID: PMC11971303 DOI: 10.1038/s41598-025-90089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/10/2025] [Indexed: 04/07/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB) infection, remains a significant public health threat. The timeliness, portability, and capacity of nanopore sequencing for diagnostics can aid in early detection and drug susceptibility testing (DST), which is crucial for effective TB control. This study synthesized current evidence on the diagnostic accuracy of the nanopore sequencing technology in detecting MTB and its DST profile. A comprehensive literature search in PubMed, Scopus, MEDLINE, Cochrane, EMBASE, Web of Science, AIM, IMEMR, IMSEAR, LILACS, WPRO, HERDIN Plus, MedRxiv, and BioRxiv was performed. Quality was assessed using the Quality Assessment of Diagnostic Accuracy Studies-2 tool. Pooled sensitivity, specificity, predictive values (PV), diagnostic odds ratio (DOR), and area under the curve (AUC) were calculated. Thirty-two studies were included; 13 addressed MTB detection only, 15 focused on DST only, and 4 examined both MTB detection and DST. No study used Flongle or PromethION. Seven studies were eligible for meta-analysis on MTB detection and five for DST; studies for MTB detection used GridION only while those for DST profile used MinION only. Our results indicate that GridION device has high sensitivity [88.61%; 95% CI (83.81-92.12%)] and specificity [93.18%; 95% CI (85.32-96.98%)], high positive predictive value [94.71%; 95% CI (89.99-97.27%)], moderately high negative predictive value [84.33%; 95% CI (72.02-91.84%)], and excellent DOR [107.23; 95% CI (35.15-327.15)] and AUC (0.932) in detecting MTB. Based on DOR and AUC, the MinION excelled in detecting pyrazinamide and rifampicin resistance; however, it underperformed in detecting isoniazid and ethambutol resistance. Additional studies will be needed to provide more precise estimates for MinION's sensitivity in detecting drug-resistance, as well as DOR in detecting resistance to pyrazinamide, streptomycin, and ofloxacin. Studies on detecting resistance to bedaquiline, pretomanid, and linezolid are lacking. Subgroup analyses suggest that overall accuracy of MTB detection tends to be higher with prospective study design and use of standards other than CSTB (Chinese national standard for diagnosing TB). Sensitivity analyses reveal that retrospective study design, use of GridION, and use of Illumina whole-genome sequencing (WGS) decrease overall accuracy in detecting any drug-resistant MTB. Findings from both types of analyses, however, should be interpreted with caution because of the low number of studies and uneven distribution of studies in each subgroup.
Collapse
Affiliation(s)
| | | | - Gail S Co
- Ateneo School of Medicine and Public Health, Pasig, 1604, Philippines
| | - John David Pilapil
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology , Kowloon, Hong Kong SAR, 999077, China
| | - Juan Ignacio Garcia
- Tuberculosis Group, Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, 78227, US
- International Center for the Advancement of Research & Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, 78227, US
| | - Blanca I Restrepo
- International Center for the Advancement of Research & Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, 78227, US
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville campus, Brownsville, TX, 7852, US
| | - Marcel Yotebieng
- International Center for the Advancement of Research & Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, 78227, US
- Division of General Internal Medicine, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, US
| | - Jordi B Torrelles
- Tuberculosis Group, Disease Intervention & Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, 78227, US.
- International Center for the Advancement of Research & Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, 78227, US.
| | - Kin Israel Notarte
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, US.
| |
Collapse
|
30
|
Wang X, Zhou H, Li D, Zhao Z, Peng K, Xu X, Wang JJ, Wang Y, Wang J, Zhang JJ, Wan SS, Shi MQ, Chen J, Ding XG, Ji FH. Molecular Targeting of Intracellular Bacteria by Homotypic Recognizing Nanovesicles for Infected Pneumonia Treatment. Biomater Res 2025; 29:0172. [PMID: 40177029 PMCID: PMC11964281 DOI: 10.34133/bmr.0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Although extensive antibiotic regimens have been implemented to address pathogen-infected pneumonia, existing strategies are constrained in their efficacy against intracellular bacteria, a prominent contributor to antibiotic resistance. In addition, the concurrent occurrence of a cytokine storm during antibiotic therapy presents a formidable obstacle in the management of pneumonia caused by pathogens. In the present study, an infection-targeting system that leverages M2-macrophage-derived vesicles [exosomes (Exos)] as vehicles to convey antibiotics (antibiotics@Exos) was developed for effective pneumonia management. The proposed system can enable antibiotics to be specifically delivered to infected macrophages in pneumonia through homotypic recognition and was found to exhibit an exceptional intracellular bactericidal effect. Moreover, M2-type vesicles exhibit a high degree of efficiency in reprogramming inflammatory macrophages toward an anti-inflammatory phenotype. As a result, the administration of antibiotics@Exos was found to substantical decrease the level of the infiltrated inflammatory cells and alleviate the inflammatory factor storm in the lungs of acute lung injury mice. This intervention resulted in the alleviation of reactive-oxygen-species-induced damage, reduction of pulmonary edema, and successful pneumonia treatment. This bioactive vesicle delivery system effectively compensates for the limitations of traditional antibiotic therapy regimens with pluralism effects, paving a new strategy for serious infectious diseases, especially acute pneumonia treatment.
Collapse
Affiliation(s)
- Xu Wang
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hao Zhou
- Department of General Surgery,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu , China
| | - Dan Li
- Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Zhe Zhao
- Suzhou Institute of Nano-Tech and Nano-Bionics, CAS Key Laboratory of Nano-Bio Interface Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology & Institute of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jia-Jia Wang
- Department of Pulmonary and Critical Care Medicine,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yang Wang
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Wang
- Department of Intensive Care Medicine,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing-Jing Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors,
Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, China
| | - Shuang-Shuang Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors,
Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, China
| | - Mai-Qing Shi
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Chen
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xian-Guang Ding
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors,
Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, China
| | - Fu-Hai Ji
- Department of Anesthesiology & Institute of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
31
|
Wang F, Zeng X, Wen J, Xian K, Jin F, Jiang S, Sun L. Identification of KIFC3 as a Colorectal Cancer Biomarker and Its Regulatory Mechanism in the Immune Microenvironment Based on Integrated Analysis of Multi-Omics Databases. Biomedicines 2025; 13:859. [PMID: 40299423 PMCID: PMC12025125 DOI: 10.3390/biomedicines13040859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Studies suggest that kinesin family (KIF) members can promote the occurrence of colorectal cancer (CRC). However, the mechanism of action has not yet been elucidated. The aim of this study was to identify CRC biomarkers associated with KIF members and to investigate their biological mechanisms in the treatment of colorectal cancer by analyzing multi-omics data. Methods: CRC-related datasets and KIF member-related genes (KIFRGs) were used. First, differentially expressed genes (DEGs) and differentially expressed methylation genes (DEMGs) in the TCGA-CRC were identified separately using different expression analyses (CRC vs. control). The intersecting genes were selected by overlapping the DEGs, DEMGs, and KIFRGs. Candidate genes were identified using survival analysis (p < 0.05). Subsequently, based on the candidate genes, biomarkers were selected by gene expression validation and survival analysis. Subsequently, functional enrichment, immune cell infiltration, and drug sensitivity analyses were performed. Single-cell analysis was utilized to perform cell annotation, and then function enrichment and pseudo-temporal analyses were performed. Results: The 12 intersecting genes were identified by overlapping 12,479 DEGs, 11,319 DEMGs, and 43 KIFRGs. The survival analysis showed that Kinesin Family Member C2 (KIFC2) and Kinesin Family Member C3 (KIFC3) had significant differences in survival (p < 0.05). Moreover, KIFC3 passed the gene expression validation and survival analysis validation (p < 0.05); thus, KIFC3 was deemed a biomarker. Subsequently, the pathways involved in KIFC3 were detected, such as the Ecm receptor intersection and chemokine signaling pathway. In addition, we found that KIFC3 was significantly positively correlated with natural killer (NK) cells (r = 0.455, p < 0.05) and NK T cells (r = 0.411, p < 0.05). Moreover, in the drug sensitivity of the CRC, the potential therapeutic benefits of AZD.2281, nilotinib, PD.173074, and shikonin were detected. Furthermore, using single-cell analysis, 16 cell clusters were annotated, and epithelial cells and M2-like macrophages were enriched in "rheumatoid arthritis". Additionally, we observed that most M1-like macrophages were present in the early stages of differentiation, whereas M2-like macrophages were predominant in the later stages of differentiation. Conclusions: This study identifies KIFC3 as a CRC biomarker through multi-omics analysis, highlighting its unique expression, survival association, immune correlations, and drug sensitivity for potential diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Fen Wang
- Department of Medical, Peking University Shenzhen Hospital, Shenzhen 518000, China; (F.W.); (F.J.)
| | - Xinxin Zeng
- Second Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou 518025, China; (X.Z.); (K.X.)
| | - Jielun Wen
- School of Medicine, Jinan University, Guangzhou 510632, China;
| | - Kexin Xian
- Second Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou 518025, China; (X.Z.); (K.X.)
| | - Feng Jin
- Department of Medical, Peking University Shenzhen Hospital, Shenzhen 518000, China; (F.W.); (F.J.)
| | - Sunfang Jiang
- Department of Health Management Centre, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liyue Sun
- Department of Health Management Centre, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
32
|
Dasharathy S, Pranay, Devadas SK, Tripathi E, Karyala P. Emerging role of deubiquitinases in modulating cancer chemoresistance. Drug Discov Today 2025; 30:104339. [PMID: 40118446 DOI: 10.1016/j.drudis.2025.104339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Chemotherapy remains a gold standard in cancer treatment by targeting the rapidly dividing cancer cells. However, chemoresistance is a major obstacle to successful cancer treatment, often leading to recurrence, metastasis, and high mortality. Deubiquitinases (DUBs), enzymes that remove ubiquitin and stabilize proteins, have been implicated in chemoresistance and can either promote therapeutic resistance or enhance sensitivity depending on their targets. In this review, we highlight the chemoresistance mechanisms of DUBs in various cancers, including breast, lung, liver, gastrointestinal, colorectal, ovarian, prostate, and blood cancers. Given these mechanisms, the development of DUB inhibitors has gained considerable attention in cancer therapeutics and combination therapies involving these inhibitors show potential to overcome drug resistance and improving treatment outcomes.
Collapse
Affiliation(s)
- Sukeerthi Dasharathy
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India
| | - Pranay
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India
| | - Santhosh K Devadas
- Department of Medical Oncology, Ramaiah Medical College and Hospital, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India
| | - Ekta Tripathi
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India.
| | - Prashanthi Karyala
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India.
| |
Collapse
|
33
|
Xiang Y, Cheng X, Li H, Xu W, Zhang W. Long Non-coding RNA FOXD2-AS1 Silencing Inhibits Malignant Behaviors of Ovarian Cancer Cells Via miR-324-3p/SOX4 Signaling Axis. Reprod Sci 2025; 32:1003-1012. [PMID: 39455487 DOI: 10.1007/s43032-024-01719-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/28/2024] [Indexed: 10/28/2024]
Abstract
It is urgent to develop new therapeutic strategies for ovarian cancer (OC). Long-noncoding RNAs (lncRNAs) have participated in multiple biological processes including tumor recurrence and progression. This study aimed to determine the effects and potential regulatory mechanism of lncRNA FOXD2-AS1 in OC progression. Levels of lncRNA FOXD2-AS1 and miR-324-3p in OC tissues and cell lines were analyzed using quantitative real-time PCR (qRT-PCR). The direct target between FOXD2-AS1 or miR-324-3p was determined using bioinformatics tools and further verified by dual-luciferase reporter assay. Cell viability, apoptosis, migration, along invasion were assessed by MTT, flow cytometry, as well as Transwell assays, respectively. In addition, the levels of miR-324-3p, PCNA, MMP9, Bax, Bcl-2, and SOX4 in OC cells were evaluated using qRT-PCR and western blot assays. We observed that lncRNA FOXD2-AS1 was up-regulated while miR-324-3p was down-regulated in OC tissues and cell lines, especially in SKOV3 cells. Moreover, miR-324-3p was a direct target of lncRNA FOXD2-AS1. Meanwhile, SOX4 interacted with miR-324-3p and was negatively regulated by miR-324-3p in SKOV3 cells. Function assays confirmed that lncRNA FOXD2-AS1 silenced depressed cell proliferation, migration, and invasion while accelerating apoptosis. These functions of lncRNA FOXD2-AS1 were attenuated by miR-324-3p inhibition. Our research demonstrated that FOXD2-AS1 silencing restrained cell growth and metastasis of OC via regulating miR-324-3p/SOX4 axis, indicating that lncRNA FOXD2-AS1 could be a novel potential therapeutic target for OC.
Collapse
Affiliation(s)
- Yun Xiang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xi Cheng
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Hong Li
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenjing Xu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Weiqiang Zhang
- Department of Gynecology and Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
34
|
Jin SK, Baek KH. Unraveling the role of deubiquitinating enzymes on cisplatin resistance in several cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189297. [PMID: 40058507 DOI: 10.1016/j.bbcan.2025.189297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
The use of platinum-based drugs in cancer treatment is one of the most common methods in chemotherapy. Especially, cisplatin induces cell death by interrupting DNA synthesis by binding to the DNA bases, thereby leading to the apoptosis via multiple pathways. However, the major hurdle in chemotherapy is drug resistance. To overcome drug resistance, the ubiquitin-proteasome system (UPS) has emerged as a potential therapeutic target. The UPS is a pivotal signaling pathway that regulates the majority of cellular proteins by attaching ubiquitin to substrates, leading to proteasomal degradation. Conversely, deubiquitinating enzymes (DUBs) remove tagged ubiquitin from the substrate and inhibit degradation, thereby maintaining proteostasis. Recently, studies have been conducted to identify the substrates of DUBs and investigated the cellular mechanisms, and now the development of therapeutics using DUB inhibitors is in clinical trials. However, the mechanism of the DUB response to cisplatin remains still unclear. In this review, we summarize the research reported on the function of DUBs responding to cisplatin.
Collapse
Affiliation(s)
- Sun-Kyu Jin
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
35
|
Anampa JD, Alvarez Soto A, Bernal AM, Acuna-Villaorduna A. Racial disparities in treatment and outcomes between Hispanic and non-Hispanic black women with triple-negative breast cancer. Breast Cancer Res Treat 2025; 210:307-317. [PMID: 39589609 DOI: 10.1007/s10549-024-07565-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is an aggressive breast cancer (BC) subtype with higher incidence and mortality rates in non-Hispanic Black (NHB) women than non-Hispanic Whites. Studies assessing disparities between NHB and Hispanic women, the two largest US racial/ethnic minorities, are lacking. This study evaluates disparities in the treatment and outcomes between NHB and Hispanic women with non-metastatic TNBC. METHODS This observational, population-based study using the SEER database included adult, female patients diagnosed with non-metastatic TNBC between 2010 and 2015 and identified as NHB or Hispanic. Logistic regression analysis was used to examine the adjusted odds of receiving breast cancer-directed treatment. Kaplan-Meier and cumulative incidence of death curves were plotted to assess overall survival (OS) and risk of breast cancer-related death, respectively. Multivariate regression analyses with Cox and Fine-Gray methods were calculated to assess factors associated with OS and breast cancer-related death, respectively. RESULTS There were 3426 Hispanic and 5419 NHB patients with non-metastatic TNBC. Hispanics had better 5-year OS relative to NHB (76% vs. 72%). No differences in the odds of receiving chemotherapy or surgery between cohorts was seen. However, the odds of undergoing breast-conserving surgery (BCS) and receiving radiation was higher in NHB than Hispanics, (OR, 1.22; 95% CI, 1.10-1.36) and (OR, 1.50; 95% CI, 1.36-1.66), respectively. Lack of radiation therapy was associated with increased BC-related death in NHB relative to Hispanics (sHR, 1.40; 95% CI, 1.19-1.65). Nevertheless, this difference was not seen when radiation was given, (sHR, 1.03; 95% CI, 0.87-1.23). CONCLUSIONS We found racial disparities in treatment and outcomes between NHB and Hispanics. NHB were more likely to receive radiation therapy and have BCS. Still, after adjusting for demographic and treatment-related factors, NHB had worse OS and BCSS relative to Hispanics. Additional research is needed to understand the drivers of these disparities.
Collapse
Affiliation(s)
- Jesus D Anampa
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, 1695 Eastchester Rd, 2nd Floor, Bronx, NY, 10461, USA.
| | - Alvaro Alvarez Soto
- Department of Medicine, Hematology/Oncology, Carole and Ray Neag Comprehensive Cancer Center, UCONN Health, Farmington, CT, USA
| | - Ana M Bernal
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, 1695 Eastchester Rd, 2nd Floor, Bronx, NY, 10461, USA
| | - Ana Acuna-Villaorduna
- Department of Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
36
|
Hu Y, Zhao Y, Zhang Y, Chen W, Zhang H, Jin X. Cell-free DNA: a promising biomarker in infectious diseases. Trends Microbiol 2025; 33:421-433. [PMID: 38997867 DOI: 10.1016/j.tim.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024]
Abstract
Infectious diseases pose serious threats to public health worldwide. Conventional diagnostic methods for infectious diseases often exhibit low sensitivity, invasiveness, and long turnaround times. User-friendly point-of-care tests are urgently needed for early diagnosis, treatment monitoring, and prognostic prediction of infectious diseases. Cell-free DNA (cfDNA), a promising non-invasive biomarker widely used in oncology and pregnancy, has shown great potential in clinical applications for diagnosing infectious diseases. Here, we discuss the most recent cfDNA research on infectious diseases from both the pathogen and host perspectives. We also discuss the technical challenges in this field and propose solutions to overcome them. Additionally, we provide an outlook on the potential of cfDNA as a diagnostic, treatment, and prognostic marker for infectious diseases.
Collapse
Affiliation(s)
- Yuxuan Hu
- BGI Research, Shenzhen 518083, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | | | - Yan Zhang
- BGI Research, Shenzhen 518083, China
| | - Weijun Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; PathoGenesis, BGI Genomics, Shenzhen 518083, China
| | | | - Xin Jin
- BGI Research, Shenzhen 518083, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China.
| |
Collapse
|
37
|
Li J, Zhou L, Hao Y, Xing C. Nanophotonic biosensors for COVID-19 detection: advances in mechanisms, methods, and design. NANOSCALE 2025; 17:7600-7616. [PMID: 40008826 DOI: 10.1039/d4nr04423a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
The growing societal impact of coronavirus disease 2019 (COVID-19) has underscored the urgent need for innovative strategies to address the ongoing challenges posed by the pandemic. While rapid therapeutic interventions remain critical for short-term mitigation, equally vital is the development of accessible and efficient diagnostic tools to curb viral transmission. In this context, optical sensing technologies have emerged as foundational tools for detection and diagnosis, owing to their rapid response, user-friendliness, and adaptability. These attributes strengthen their indispensable role in identifying severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19. This review systematically outlines the structural components of SARS-CoV-2 virions and their respective biological functions, classifies optical biosensors according to their underlying principles and evaluates the advantages and limitations of each methodology in real-world diagnostic applications. By addressing current detection challenges, these optical platforms not only enhance our capacity to manage SARS-CoV-2 but also establish a framework for deploying optical sensing technologies in future pandemic scenarios.
Collapse
Affiliation(s)
- Jiawei Li
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, People's Republic of China.
| | - Linyan Zhou
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, People's Republic of China.
| | - Yabin Hao
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, People's Republic of China.
| | - Chenyang Xing
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, People's Republic of China.
- State Key Laboratory of Radio Frequency Heterogeneous Integration, Shenzhen University, Shenzhen 518060, People's Republic of China
| |
Collapse
|
38
|
Lei P, Yan YH, Jiang Y, Wang Y, Xiong R, Deng J, Zhang H, Yang Z, Zhang W, Wu JW, Liu W, Lei H, Li GB, Yang L. Discovery of New Azaindole Metallo-Deubiquitinase CSN5 Inhibitors. J Med Chem 2025; 68:6748-6765. [PMID: 40053484 DOI: 10.1021/acs.jmedchem.5c00184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Abstract
CSN5 is responsible for the deneddylation of cullin-RING E3 ubiquitin ligases and is closely linked to the development of various cancers. We previously developed a noncatalytic activity assay platform using novel fluorescent probes derived from azaindole inhibitors, which also highlighted the potential for further structural optimization of azaindoles. Herein, we report a series of new 4-NH-substituted azaindole derivatives, some of which showed nanomolar activity against the CSN5 subunit. Cellular assays revealed that the new azaindoles increase the cullin 1 neddylation in cancer cells. Importantly, they exhibit synergistic anticancer effects in combination with poly(ADP-ribose) polymerase inhibitors through increasing DNA damage. This work presents a new lead compound and a potential combination strategy for drug discovery targeting CSN5.
Collapse
Affiliation(s)
- Pengcheng Lei
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Yu-Hang Yan
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yingying Jiang
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Yanjun Wang
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Rui Xiong
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Jianlin Deng
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Hang Zhang
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Zhiwen Yang
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Weifeng Zhang
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Jing-Wei Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wenyi Liu
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Hui Lei
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Guo-Bo Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lingling Yang
- College of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| |
Collapse
|
39
|
Wakle KS, Karwa PN, Sakle NS. Investigating Vitamin D 3's anticancer mechanisms in MCF-7 cells: a network pharmacology and omics technology approach. Mol Divers 2025:10.1007/s11030-025-11156-z. [PMID: 40146431 DOI: 10.1007/s11030-025-11156-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025]
Abstract
Breast cancer is one of the leading reasons of mortality due to cancer globally. Estrogen receptor-positive (ER +) breast cancer being a significant subtype. The therapeutic potential of Vitamin D3 in cancer treatment has gained attention due to its ability to modulate key molecular targets and signaling pathways. This study investigates the anticancer mechanisms of Vitamin D3 in MCF-7 breast cancer cells using network pharmacology and omics technology approach. Utilizing protein-protein interaction (PPI) networks, we identified several critical protein targets involved in breast cancer progression, including ESR1, ESR2, PGR, IGF1R, and KDR. Pathway enrichment analyses highlighted Vitamin D3's impact on pivotal signaling pathways such as the PI3K/Akt pathway, estrogen receptor signaling, and apoptosis regulation. In vitro studies showed that Vitamin D3 significantly inhibited cell proliferation in MCF-7 cells. It also induced apoptosis and disrupted mitochondrial function. Flow cytometry analysis demonstrated a dose-dependent increase in apoptotic cell death and S-phase cell cycle arrest. Confocal imaging and mitochondrial membrane potential assays further supported the findings, indicating mitochondrial dysfunction and chromatin condensation. Additionally, gene expression analysis in breast invasive carcinoma tissues confirmed the relevance of ESR1 and PGR in hormone receptor-positive breast cancer. Histopathological studies on DMBA-induced mammary carcinoma revealed Vitamin D3's protective effects, reducing tumor malignancy severity through anti-proliferative and pro-apoptotic actions. These findings provide strong evidence for Vitamin D3's potential as a multi-targeted therapeutic agent in breast cancer, suggesting further investigation into its clinical applications and combination strategies with existing therapies as an adjunct or alternative in the treatment.
Collapse
Affiliation(s)
- Komal S Wakle
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, 431001, India
| | - Pawan N Karwa
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, India
| | - Nikhil S Sakle
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, 431001, India.
| |
Collapse
|
40
|
Gu XY, Yang JL, Lai R, Zhou ZJ, Tang D, Hu L, Zhao LJ. Impact of lactate on immune cell function in the tumor microenvironment: mechanisms and therapeutic perspectives. Front Immunol 2025; 16:1563303. [PMID: 40207222 PMCID: PMC11979165 DOI: 10.3389/fimmu.2025.1563303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Lactate has emerged as a key regulator in the tumor microenvironment (TME), influencing both tumor progression and immune dynamics. As a byproduct of aerobic glycolysis, lactate satisfies the metabolic needs of proliferating tumor cells while reshaping the TME to facilitate immune evasion. Elevated lactate levels inhibit effector immune cells such as CD8+ T and natural killer cells, while supporting immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells, thus fostering an immunosuppressive environment. Lactate promotes epigenetic reprogramming, stabilizes hypoxia-inducible factor-1α, and activates nuclear factor kappa B, leading to further immunological dysfunction. In this review, we examined the role of lactate in metabolic reprogramming, immune suppression, and treatment resistance. We also discuss promising therapeutic strategies targeting lactate metabolism, including lactate dehydrogenase inhibitors, monocarboxylate transporter inhibitors, and TME neutralization methods, all of which can restore immune function and enhance immunotherapy outcomes. By highlighting recent advances, this review provides a theoretical foundation for integrating lactate-targeted therapies into clinical practice. We also highlight the potential synergy between these therapies and current immunotherapeutic strategies, providing new avenues for addressing TME-related challenges and improving outcomes for patients with cancer.
Collapse
Affiliation(s)
- Xuan-Yu Gu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia-Li Yang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Rui Lai
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zheng-Jun Zhou
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dan Tang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Hepatobiliary and Pancreatic Surgery, Suzhou Medical College of Soochow University, Suzhou, China
| | - Long Hu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | - Li-Jin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
41
|
Nikbakht F, Heidarian Miri H, Mosafarkhani E, Sharifjafari F, Taghipour A. Role of COVID-19 infection status on the prediction of future infection: Immunity or susceptibility. PLoS One 2025; 20:e0317959. [PMID: 40138353 PMCID: PMC11940750 DOI: 10.1371/journal.pone.0317959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND COVID-19 has rapidly spread around the world, and the duration of protective immunity against the virus remains unknown. Evidence suggests that patients with a confirmed COVID-19 infection may experience reinfection. The aim of this study is to determine the relationship between COVID-19 reinfection and previous infection history in the population covered by Mashhad University of Medical Sciences. METHODS This population-based, historical cohort study included all individuals with health records at the health service centers of Mashhad University of Medical Sciences who underwent PCR testing during the study period (April 1, 2020, up to February 19, 2022). The data were analyzed by calculating the infection rate in both PCR-positive and negative individuals, and estimating the adjusted rate ratio using Poisson regression. RESULTS The results of this study in the entire population showed that the incidence rate in people with a history of primary COVID-19 infection was 13% higher than that in people who had no history of this disease. However, in the group that received the vaccine prior to the first PCR test, the incidence rate was lower among individuals with a positive first test result (IRR = 0.71) compared to those with a negative first test result. CONCLUSION The study reveals that prior COVID-19 infection does not ensure immunity and may increase the risk of reinfection, particularly among men and younger individuals. Vaccination appears to complicate this dynamic, as those with multiple vaccine doses showed higher reinfection rates compared to those with fewer doses. These findings highlight the need for ongoing research and tailored public health strategies to address the complexities of COVID-19 immunity and reinfection.
Collapse
Affiliation(s)
- Fateme Nikbakht
- Department of Epidemiology, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ehsan Mosafarkhani
- Department of Epidemiology, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
- Management and Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Taghipour
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
42
|
Chang L, Qin C, Chu Y, Guan M, Deng X. Migrasome-Related Genes as Potential Prognosis and Immunotherapy Response Predictors for Colorectal Cancer. Biomedicines 2025; 13:799. [PMID: 40299331 PMCID: PMC12024535 DOI: 10.3390/biomedicines13040799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Studies highlight the role of migrasomes as mediators of intercellular communication and signaling, critical in influencing tumorigenesis and progression. Yet migrasome-related genes and their potential role in colorectal cancer prognosis remain unexplored. Methods: Differentially expressed gene set A (DEG set A) was identified in the TCGA-CRC dataset, and Weighted Gene Co-expression Network Analysis (WGCNA) was performed to identify the most important modules associated with migrasome-related gene (MRG) scores. Single-cell RNA-seq dataset GSE231559 DEG set B was determined. Candidate migrasome-related genes were filtered by intersecting DGE set A, key module genes, and DEG set B. Prognostic genes were subsequently screened through regression analysis, and a risk model was developed. Patients with CRC in the TCGA cohort were stratified into high- and low-risk groups based on the optimal cutoff of the risk score. Immunotherapy response-related analyses were then performed. Finally, cell-to-cell communication analysis was carried out for key cells identified based on prognostic gene expression analysis in annotated cells. Results: The six candidate migrasome-related genes were identified through the overlap of 5158 DEG set A, 1960 key module genes, and 146 DEG set B. Further screening led to the selection of T1MP1, CXCL8, and MGP as potential prognostic biomarkers. Immune-related analysis indicated that the high-risk group exhibited a better response to immunotherapy. Notably, the prognostic genes showed elevated expression levels in monocytes and tissue stem cells, thereby designating them as key cell types. Conclusions: We conducted bioinformatic analysis of migrasome-related genes and identified significant involvement of T1MP1, CXCL8, and MGP in influencing CRC prognosis and immunotherapy response. Our research provides novel insights into the role of migrasomes in CRC biology.
Collapse
Affiliation(s)
- Lu Chang
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| | - Chao Qin
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| | - Yimin Chu
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200050, China;
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| | - Xuan Deng
- Department of Laboratory Medicine, Huashan Hospital Fudan University, Shanghai 200040, China; (L.C.); (C.Q.)
| |
Collapse
|
43
|
Zhang H, Zhang J, Zhu K, Li S, Liu J, Guan B, Zhang H, Chen C, Liu Y. Identification and characterization of mitochondrial autophagy-related genes in osteosarcoma and predicting clinical prognosis. Sci Rep 2025; 15:10158. [PMID: 40128298 PMCID: PMC11933398 DOI: 10.1038/s41598-025-95173-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/19/2025] [Indexed: 03/26/2025] Open
Abstract
Osteosarcoma (OS), the most prevalent primary malignant bone tumor, is characterized by a poor prognosis and high metastatic potential. Mitochondrial autophagy has been implicated in cancer suppression. This study aimed to identify prognostic genes associated with mitochondrial autophagy in OS. Public datasets, including TARGET-OS, GSE99671, and GSE21257, were retrieved for analysis. Differentially expressed genes (DEGs1) between OS and normal samples were identified from GSE99671. Single-sample Gene Set Enrichment Analysis (ssGSEA) was applied to quantify the enrichment scores of 29 mitochondrial autophagy-related genes (MARGs) in OS samples from TARGET-OS, categorizing them into high- and low-score groups to extract DEGs2. The intersection of DEGs1 and DEGs2 yielded mitochondrial autophagy-associated differentially expressed genes (MDGs). Prognostic genes were subsequently screened through a multi-step regression analysis, and a risk score was computed. TARGET-OS samples were stratified into high- and low-risk groups based on the optimal cutoff value of the risk score. GSEA was conducted between the two risk groups. Additionally, associations between prognostic genes and the immune microenvironment were explored. A total of 31 MDGs were identified from the overlap of 3,207 DEGs1 and 622 DEGs2. Five prognostic genes-KLK2, NRXN1, HES5, OR2W3, and HS3ST4-were further selected. Kaplan-Meier survival analysis indicated significantly reduced survival in the high-risk group. GSEA revealed enrichment in ABC transporter activity and glycolysis/gluconeogenesis pathways. Immunoanalysis demonstrated significant differences in 11 immune cell populations and three immune functions between risk groups, notably myeloid-derived suppressor cells (MDSCs) and Type 1 T helper cells. HS3ST4 exhibited the strongest positive correlation with macrophages, whereas NRXN1 showed the most pronounced negative correlation with memory B cells. Expressions of HAVCR2 and PDCD1LG2 were elevated in the low-risk group. Functional analysis indicated significant differences in dysfunction patterns between risk groups. This study identified five mitochondrial autophagy-related prognostic genes and constructed a risk model, offering novel insights into OS diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jingyu Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Kai Zhu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Shuang Li
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jinwei Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Boya Guan
- Department of Pharmacy, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Hong Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Changbao Chen
- Department of Spinal Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yancheng Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
| |
Collapse
|
44
|
Liu Z, Ou Y, He X, Yuan T, Li M, Long Y, Li Y, Tan Y. Guardians of the Lung: The Multifaceted Roles of Macrophages in Cancer and Infectious Disease. DNA Cell Biol 2025. [PMID: 40106386 DOI: 10.1089/dna.2024.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
The lung as an organ that is fully exposed to the external environment for extended periods, comes into contact with numerous inhaled microorganisms. Lung macrophages are crucial for maintaining lung immunity and operate primarily through signaling pathways such as toll-like receptor 4 and nuclear factor-κB pathways. These macrophages constitute a diverse population with significant plasticity, exhibiting different phenotypes and functions on the basis of their origin, tissue residence, and environmental factors. During lung homeostasis, they are involved in the clearance of inhaled particles, cellular remnants, and even participate in metabolic processes. In disease states, lung macrophages transition from the inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. These distinct phenotypes have varying transcriptional profiles and serve different functions, from combating pathogens to repairing inflammation-induced damage. However, macrophages can also exacerbate lung injury during prolonged inflammation or exposure to antigens. In this review, we delve into the diverse roles of pulmonary macrophages the realms in homeostasis, pneumonia, tuberculosis, and lung tumors.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Zhuzhou, China
| | - Yangjing Ou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xiaojin He
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Ting Yuan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Miao Li
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yunzhu Long
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yingzheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
45
|
Dong J, Li P, Liu Q, Liu Q, Wang C, Zhao X, Hu X. A flexible transoral swab sampling robot system with visual-tactile fusion approach. Front Robot AI 2025; 12:1520374. [PMID: 40177224 PMCID: PMC11961991 DOI: 10.3389/frobt.2025.1520374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
A significant number of individuals have been affected by pandemic diseases, such as COVID-19 and seasonal influenza. Nucleic acid testing is a common method for identifying infected patients. However, manual sampling methods require the involvement of numerous healthcare professionals. To address this challenge, we propose a novel transoral swab sampling robot designed to autonomously perform nucleic acid sampling using a visual-tactile fusion approach. The robot comprises a series-parallel hybrid flexible mechanism for precise distal posture adjustment and a visual-tactile perception module for navigation within the subject's oral cavity. The series-parallel hybrid mechanism, driven by flexible shafts, enables omnidirectional bending through coordinated movement of the two segments of the bendable joint. The visual-tactile perception module incorporates a camera to capture oral images of the subject and recognize the nucleic acid sampling point using a deep learning method. Additionally, a force sensor positioned at the distal end of the robot provides feedback on contact force as the swab is inserted into the subject's oral cavity. The sampling robot is capable of autonomously performing transoral swab sampling while navigating using the visual-tactile perception algorithm. Preliminary experimental trials indicate that the designed robot system is feasible, safe, and accurate for sample collection from subjects.
Collapse
Affiliation(s)
- Jiaxiang Dong
- School of Mechanical and Automotive Engineering, South China University of Technology, Guangzhou, China
| | - Peng Li
- School of Mechanical and Electrical Engineering and Automation, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Quanquan Liu
- Artificial Intelligence Research Institute & Guangdong-Hong Kong-Macao Joint Laboratory, Shenzhen MSU-BIT University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qi Liu
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Chunbao Wang
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- School of Mechanical and Electrical Engineering, Guangdong University of Science and Technology, Dongguan, China
| | - Xuezhi Zhao
- School of Mechanical and Automotive Engineering, South China University of Technology, Guangzhou, China
| | - Xiping Hu
- Artificial Intelligence Research Institute & Guangdong-Hong Kong-Macao Joint Laboratory, Shenzhen MSU-BIT University, Shenzhen, China
| |
Collapse
|
46
|
Jin Y, Gu W. Prognostic and clinicopathological value of the controlling nutritional status score in patients with multiple myeloma: a meta-analysis. Front Oncol 2025; 15:1517223. [PMID: 40171257 PMCID: PMC11959075 DOI: 10.3389/fonc.2025.1517223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Background The effect of the controlling nutritional status (CONUT) score on forecasting multiple myeloma (MM) prognosis is previously analyzed, whereas the results remained inconsistent. The present meta-analysis focused on identifying the exact function of CONUT in forecasting MM prognosis. Methods Web of Science, PubMed, Embase, CNKI, and Cochrane Library were comprehensively searched between inception and 1 February 2025. The effect of CONUT on forecasting MM overall survival (OS) and progression-free survival (PFS) was determined by computing pooled hazard ratios (HRs) together with 95% confidence intervals (CIs). Results There were nine studies with 1,176 patients being recruited into the present work. As indicated by our pooled data, elevated CONUT was related to the dismal OS (HR = 1.87, 95% CI = 1.37-2.54, p < 0.001) of patients with MM. Nonetheless, CONUT was not significantly related to PFS (HR = 1.33, 95% CI = 0.81-2.19, p = 0.254) of MM. Furthermore, higher CONUT score showed a significant relationship to bone marrow plasma cells >30% (OR = 2.30, 95% CI = 1.32-3.99, p = 0.003). On the other hand, CONUT was not markedly correlated with gender (OR = 2.68, 95% CI = 0.81-8.82, p = 0.105), ISS stage (OR = 1.28, 95% CI = 0.94-1.75, p = 0.119), or ECOG PS (OR = 1.30, 95% CI = 0.84-2.01, p = 0.234) of MM. Conclusion Collectively, according to our results in this meta-analysis, higher CONUT score is markedly related to dismal OS, but not PFS in patients with MM. CONUT score can be used as a candidate marker used to predict MM prognosis in the clinic in the future.
Collapse
Affiliation(s)
| | - Wenfei Gu
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou
University, Huzhou, Zhejiang, China
| |
Collapse
|
47
|
Jin Y, Liang Y, Wu B, Wu S, Liu X, Zhou F. Establishment of CD34 + hematopoietic stem cell-derived xenograft model of hyperleukocytic acute myeloid leukemia. BMC Cancer 2025; 25:499. [PMID: 40102796 PMCID: PMC11917077 DOI: 10.1186/s12885-025-13907-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Hyperleukocytic acute myeloid leukemia (HLL) is marked by high early mortality and presents significant therapeutic challenges. Research on HLL is still in its infancy, and comprehensive development of patient-derived xenograft (PDX) models, especially CD34 + hematopoietic stem cell-derived models, remains limited. METHODS We evaluated the establishment of the HLL model through blood examinations, smear analysis, bone marrow biopsy, flow cytometry, and mutation analysis. Correlation between survival times in mice and patients was assessed using linear regression. RESULTS In the HLL PDX mouse model, leukocyte counts could reach up to 37.35^10⁹/L, and immunophenotyping revealed the presence of hCD45+, hCD15+, and hCD33 + cells in both peripheral blood (PB) and bone marrow (BM) following inoculation with PB-derived cells for the establishment of the HLL PDX model. Similar results were observed with cells derived from the patient's BM. In the CD34 + hematopoietic stem cell-derived xenograft model, extensive infiltration of CD34 + cells into the BM, liver, and spleen was observed. Additionally, human WT1 and NRAS mutations were identified in the liver, spleen, and BM of the mice. A comparative analysis of multiple experiments revealed that shorter survival times were observed in mice receiving a higher irradiation dose of 2.5 Gy and a greater number of cells derived from PB. Additionally, shorter survival times were observed in model mice injected with cells carrying NRAS, DNMT3A, FLT3, or NPM1 gene mutations. Correlation analysis indicated that the survival times of the mice were significantly associated with the survival status of the patients. CONCLUSIONS We successfully established a CD34 + hematopoietic stem cell-derived xenograft model of HLL, providing a valuable tool for mechanistic research, drug screening, individualized therapy, and precision medicine. TRIAL REGISTRATION Not application.
Collapse
Affiliation(s)
- Yanxia Jin
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
- College of Life Sciences, Hubei Normal University, Huangshi, 435002, Hubei, China
| | - Yuxing Liang
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Balu Wu
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Sanyun Wu
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiaoyan Liu
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China.
| | - Fuling Zhou
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China.
- Research Center for Lifeorgdivision Health, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
48
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
49
|
Peng W, Shi M, Hu B, Jia J, Li X, Wang N, Man S, Ye S, Ma L. Nanotechnology-leveraged CRISPR/Cas systems: icebreaking in trace cancer-related nucleic acids biosensing. Mol Cancer 2025; 24:78. [PMID: 40087758 PMCID: PMC11908094 DOI: 10.1186/s12943-024-02222-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/31/2024] [Indexed: 03/17/2025] Open
Abstract
As promising noninvasive biomarkers, nucleic acids provide great potential to innovate cancer early detection methods and promote subsequent diagnosis to improve the survival rates of patient. Accurate, straightforward and sensitive detection of such nucleic acid-based cancer biomarkers in complex biological samples holds significant clinical importance. However, the low abundance creates huge challenges for their routine detection. As the next-generation diagnostic tool, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein (Cas) with their high programmability, sensitivity, fidelity, single-base resolution, and precise nucleic acid positioning capabilities are extremely attractive for trace nucleic acid-based cancer biomarkers (NABCBs), permitting rapid, ultra-sensitive and specific detection. More importantly, by combing with nanotechnology, it can solve the long-lasting problems of poor sensitivity, accuracy and simplicity, as well as to achieve integrated miniaturization and portable point-of-care testing (POCT) detection. However, existing literature lacks specific emphasis on this topic. Thus, we intend to propose a timely one for the readers. This review will bridge this gap by providing insights for CRISPR/Cas-based nano-biosensing development and highlighting the current state-of-art, challenges, and prospects. We expect that it can provide better understanding and valuable insights for trace NABCBs detection, thereby facilitating advancements in early cancer screening/detection/diagnostics and win practical applications in the foreseeable future.
Collapse
Affiliation(s)
- Weipan Peng
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Mengting Shi
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Bin Hu
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Jingyu Jia
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xinyue Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Nan Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Shengying Ye
- Pharmacy Department, The 983th Hospital of The Joint Logistics Support Force of The Chinese People's Liberation Army, Tianjin, China.
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| |
Collapse
|
50
|
Trivadila T, Iswantini D, Rahminiwati M, Rafi M, Salsabila AP, Sianipar RNR, Indariani S, Murni A. Herbal Immunostimulants and Their Phytochemicals: Exploring Morinda citrifolia, Echinacea purpurea, and Phyllanthus niruri. PLANTS (BASEL, SWITZERLAND) 2025; 14:897. [PMID: 40265854 PMCID: PMC11945065 DOI: 10.3390/plants14060897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025]
Abstract
The rising prevalence of infectious diseases and immune-related disorders underscores the need for effective and accessible therapeutic solutions. Herbal immunostimulants derived from medicinal plants offer promising alternatives, enhancing immune responses with lower toxicity and fewer side effects than synthetic drugs. This review explores the immunostimulatory potential of Morinda citrifolia, Echinacea purpurea, and Phyllanthus niruri, focusing on their bioactive compounds, mechanisms of action, and therapeutic relevance. These plants modulate innate and adaptive immune responses by activating macrophages, dendritic cells, and lymphocytes while regulating cytokine production to maintain immune homeostasis. Their immunomodulatory effects are linked to key signaling pathways, including NF-κB, MAPK, and JAK/STAT. In vitro and in vivo studies highlight their potential to strengthen immune responses and control inflammation, making them promising candidates for managing infectious and immune-related diseases. However, further research is needed to standardize formulations, determine optimal dosages, and validate safety and efficacy in clinical settings. Addressing these gaps will support the integration of herbal immunostimulants into evidence-based healthcare as sustainable and accessible immune-enhancing strategies.
Collapse
Affiliation(s)
- Trivadila Trivadila
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Dyah Iswantini
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Min Rahminiwati
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
- School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, West Java, Indonesia
| | - Mohamad Rafi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Adisa Putri Salsabila
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
| | - Rut Novalia Rahmawati Sianipar
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
| | - Susi Indariani
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Anggia Murni
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| |
Collapse
|