1
|
Li A, Yi Z, Ma C, Sun B, Zhao L, Cheng X, Hui L, Xia Y. Innate immune recognition in hepatitis B virus infection. Virulence 2025; 16:2492371. [PMID: 40253712 PMCID: PMC12013422 DOI: 10.1080/21505594.2025.2492371] [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: 11/28/2024] [Revised: 03/19/2025] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
Hepatitis B virus (HBV) remains a major global public health challenge, with approximately 254 million individuals chronically infected worldwide. The interaction between HBV and the innate immune system has garnered significant attention within the scientific community, with numerous studies exploring this relationship over the past several decades. While some research suggests that HBV infection activates the host's innate immune response, other studies indicate that HBV suppresses innate immune signaling pathways. These conflicting findings underscore the complexity of the HBV-innate immunity interaction, which remains inadequately understood. This review aims to clarify this interplay by examining it from three perspectives: (a) studies showing HBV activation of innate immunity; (b) evidence suggesting HBV suppression of innate immunity; and (c) findings that support HBV's role as a stealth virus. By synthesizing these perspectives, we aim to deepen the understanding of virus-host interactions that are crucial to HBV persistence and immune evasion, with potential implications for developing new therapeutic strategies for chronic HBV infection.
Collapse
Affiliation(s)
- Aixin Li
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Zhengjun Yi
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Chunqiang Ma
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Bangyao Sun
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Li Zhao
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Xiaoming Cheng
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, China
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
- Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Lixia Hui
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
- Pingyuan Laboratory, Henan, China
| |
Collapse
|
2
|
Makkar S, Rana N, Priyadarshi N, Bajaj G, Kumar S, Singhal NK. Unravelling the therapeutic properties of aptamer-modified exosome nanocomposite. Adv Colloid Interface Sci 2025; 342:103517. [PMID: 40245577 DOI: 10.1016/j.cis.2025.103517] [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: 12/30/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Exosomes are naturally occurring nanocarriers derived from various cells. In recent years, they have attained significant attention for their potential in precise drug delivery and therapeutic applications. Exosomes exhibit several advantages, remarkably improved stability, bioavailability, and delivery efficiency, which are further augmented by integration with nanomaterials. Functionalizing the aptamer and nanomaterial on the exosomal surface significantly improves the binding affinity and specificity. Here in this review, we examine the synergistic therapeutic effect of exosome-nanomaterial-aptamer conjugate with particular attention to their uses in cancer therapy, bone fracture regeneration, wound healing, etc. Recent advances in the field demonstrated that the amalgamation of different nanomaterials, aptamers, and exosomes has proven to be a transformative approach in the field of therapeutics. Here in the nanocomposite, the aptamer is exclusively used as a recognition molecule to provide specificity to the target cells. Exosomes serve as biocompatible nanocarriers, and different nanomaterials (AuNPs, AuNRs, SiNPs, Graphene, etc.) complement the therapeutic efficiency by PTT/PDT/ROS generation/SO generation, etc. Briefly, the above-mentioned nanocomposite serves as the perfect therapeutic agent by utilizing the exosome's biocompatibility, aptamer's high affinity and nanomaterial's multifunctionality. Furthermore, the challenges and limitations of this nanocomposite have been discussed, along with its prospects in clinical practices.
Collapse
Affiliation(s)
- Simran Makkar
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Niket Rana
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Nitesh Priyadarshi
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Geetika Bajaj
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Sandeep Kumar
- Department of Physics, Punjab Engineering College (Deemed to be University), Chandigarh 160012, India
| | - Nitin Kumar Singhal
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India.
| |
Collapse
|
3
|
Yin Z, Huang D, Kuhn EMA, Moriarty TF, Li G, Wang X. Unraveling persistent bacteria: Formation, niches, and eradication strategies. Microbiol Res 2025; 297:128189. [PMID: 40311456 DOI: 10.1016/j.micres.2025.128189] [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: 03/12/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Persistent bacteria (persisters) are phenotypic variants that emerge either randomly or in response to a range of adverse environmental conditions. Persistence represents a state whereby a subpopulation of microorganisms can spontaneously enter a "dormant" state in response to environmental factors, while simultaneously exhibiting elevated tolerance to antimicrobial agents. This review provides the current definition of bacterial persistence and summarizes the mechanisms of persisters formation as well as the various niches of bacterial persistence encountered in clinical practice. Strategies targeting persisters are outlined, including but not limited to direct killing, awakening of persistent bacteria, combined clearance, and inhibition of persistence formation, and we conclude by proposing challenges and solutions for addressing bacterial persistence in current clinical practice.
Collapse
Affiliation(s)
- Zibo Yin
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China
| | - Diandian Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China
| | | | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China.
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China.
| |
Collapse
|
4
|
Feng J, Xiong M, Qiao Y, Wang P, Lin X, Kang L, Duan N, Wang Z, Wu S. Ultrasensitive point-of-care testing of salmonella in lettuce and milk samples using a recombinase polymerase amplification-based colorimetric/fluorescent dual-readout lateral flow assay. Food Chem 2025; 481:144058. [PMID: 40163984 DOI: 10.1016/j.foodchem.2025.144058] [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: 11/05/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
A portable dual-mode RPA-based LFA sensor was developed for Salmonella detection in lettuce and milk samples. The fimY gene of Salmonella was chosen as specific conserved sequences, their corresponding primers based on RPA conditions were designed and optimized, and respectively labeled with Biotin and FAM groups at 5' end, then applied for efficient fimY gene fragments enrichment via RPA. Sandwich construction of streptavidin-RPA products-AuNPs@FAM antibody was generated on T-line, showing positive results within 10 min, colorimetric-fluorescent signals were captured by a smartphone and converted into gray intensities and fluorescence intensities, further enabling the quantification of Salmonella with detection limits of 10 cfu/mL and 1 cfu/mL for colorimetric and fluorescent modes, respectively, over a linear range of 101-106 cfu/mL. The proposed strategy provided a valuable reference for Salmonella one-site detection, which would be utilized as a universal method for other food-borne pathogens detection by matching specific conserved sequences and primer designs.
Collapse
Affiliation(s)
- Jiaqi Feng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Mengyuan Xiong
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yongsheng Qiao
- Key Laboratory of FSMP Risk Identification and Key Analysis Technology for Jiangsu Market Regulation, Taizhou Institute for Food Control, Taizhou, Jiangsu Province 225300, China
| | - Ping Wang
- Key Laboratory of FSMP Risk Identification and Key Analysis Technology for Jiangsu Market Regulation, Taizhou Institute for Food Control, Taizhou, Jiangsu Province 225300, China
| | - Xianfeng Lin
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lixin Kang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Nuo Duan
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhouping Wang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shijia Wu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
5
|
Chai H, Yao S, Gao Y, Hu Q, Su W. Developments in the connection between epithelial‑mesenchymal transition and endoplasmic reticulum stress (Review). Int J Mol Med 2025; 56:102. [PMID: 40341397 PMCID: PMC12081031 DOI: 10.3892/ijmm.2025.5543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/22/2025] [Indexed: 05/10/2025] Open
Abstract
Endoplasmic reticulum stress (ERS) and epithelial‑mesenchymal transition (EMT) have important roles in fibrosis and tumour development. Moderate ERS activates cellular defence mechanisms in response to noxious stimuli; however, sustained or overly strong ERS induces apoptosis. In this disease process, EMT induces epithelial cells to acquire the ability to migrate and invade. Reportedly, ERS directly or indirectly regulates EMT processes through multiple mechanisms (such as key transcription factors, signalling pathways, ferroptosis, autophagy and oxidative stress), and both processes form a complex network of interactions. Given the critical roles of ERS and EMT in disease, targeted intervention of these two processes has emerged as a potential therapeutic strategy. In the present study, the molecular interaction mechanism of ERS and EMT was systematically explored, research progress in fibrotic and neoplastic diseases was reviewed and the potential application prospects of related targeted therapies were examined, which may provide new ideas for the development of drugs to reverse fibrosis and treat tumours.
Collapse
Affiliation(s)
- Hongyu Chai
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ya Gao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qian Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Wei Su
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
6
|
Li S, Zhang J, Wei W, Zhang Z, Huang W, Xia L. The important role of myeloid-derived suppressor cells: From hepatitis to liver cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189329. [PMID: 40262654 DOI: 10.1016/j.bbcan.2025.189329] [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: 09/26/2024] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Liver homeostasis is coordinated by crosstalk between resident and infiltrating inflammatory cells. Liver disease creates a dynamic inflammatory microenvironment characterized by aberrant metabolism and continuous hepatic regeneration, making it an important risk factor for hepatocellular carcinoma (HCC) as well as liver failure. Recent studies have revealed a critical heterogeneous population of myeloid-derived suppressor cells (MDSCs), which influence liver disease progression and malignancy by dynamically regulating the immune microenvironment. MDSCs play an important role in preventing excessive immune responses in the liver. However, MDSCs are also associated with the promotion of liver injury and liver cancer progression. The plasticity of MDSCs in liver disease is a unique challenge for therapeutic intervention strategies and requires a deeper understanding of the underlying mechanisms. Here, we review the role of MDSCs in the establishment and progression of liver disease and highlight the evidence for MDSCs as a priority target for current and future therapeutic strategies. We explore the fate of MDSCs from hepatitis to liver cancer, providing recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wang Wei
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhicheng Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
7
|
Meng XZ, Duan Y, Bai Y, Zhang W, Zhang C, Wang KJ, Chen F. Litopeidin 28-51, a novel antimicrobial peptide from Litopenaeus vannamei, combats white spot syndrome virus infection through direct virus lysis and immunomodulatory effects. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110243. [PMID: 40032210 DOI: 10.1016/j.fsi.2025.110243] [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: 01/08/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
White spot syndrome virus (WSSV) poses a critical threat to crustacean aquaculture, particularly shrimp, causing widespread pandemics. In crustaceans, hemocytes function as a key component of the innate immune system and play a pivotal role in both cellular and humoral immune responses by producing various immune factors, such as antimicrobial peptides (AMPs), to defend against pathogenic microorganisms. In this study, an uncharacterized functional gene named Litopeidin was identified in Pacific white shrimp (Litopenaeus vannamei). It exhibited heightened expression in hemocytes and demonstrated a significant response to WSSV infection. Further, a truncated peptide, Litopeidin28-51, derived from this gene, was characterized and identified as a novel AMP with robust antibacterial and antifungal properties, especially against common aquatic pathogens, including Vibrio spp. Moreover, Litopeidin28-51 significantly suppressed the expression of viral genes (IE1 and VP28, WSSV replication-related genes) and the VP28 protein, as well as reduced viral copy numbers in hematopoietic tissue (Hpt) cells following WSSV infection. Mechanistic studies revealed that Litopeidin28-51 exhibited a direct virucidal effect on WSSV and significantly upregulated immune-related gene expression (including Relish, ALF, Crustin, and LYZ1) in Hpt cells. Notably, in Cherax quadricarinatus and L. vannamei, either co- or pre-treatment with Litopeidin28-51 markedly reduced animal mortality and viral replication in tissues. Collectively, the findings suggest that Litopeidin28-51, a newly identified AMP with potent antibacterial activity, effectively inhibits WSSV replication by disrupting the viral envelope and regulating the cellular antiviral responses, making it a promising candidate for developing anti-infective agents or immunostimulants in aquaculture.
Collapse
Affiliation(s)
- Xin-Zhan Meng
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Yingyi Duan
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Yuqi Bai
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Weibin Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Chang Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China; Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China; Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
8
|
Bagheri AM, Mirzahashemi M, Salarpour S, Dehghnnoudeh Y, Banat IM, Ohadi M, Dehghannoudeh G. Potential anti-aging applications of microbial-derived surfactantsin cosmetic formulations. Crit Rev Biotechnol 2025; 45:766-787. [PMID: 39294002 DOI: 10.1080/07388551.2024.2393420] [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: 12/16/2023] [Revised: 07/14/2024] [Accepted: 07/30/2024] [Indexed: 09/20/2024]
Abstract
The skin aging process is a complex interaction of genetic, epigenetic, and environmental factors, such as chemical pollution and UV radiation. There is growing evidence that biosurfactants, especially those of microbial origin, have distinct age-supportive effects through different mechanisms, such as stimulation of fibroblast growth, high antioxidant capacities, and favorable anti-inflammatory properties. With a growing financial contribution of more than 15 m€per year, microbial surfactants (MSs) display unique biological effects on the skin including improved cell mobility, better nutrient access, and facilitated cellular growth under harsh conditions. Their biodegradable nature, unusual surface activity, good safety profile and tolerance to high temperature and pH variations widen their potential spectrum in biomedical and pharmaceutical applications. MSs typically have lower critical micelle concentration (CMC) levels than chemical surfactants enhancing their effectiveness. As natural surfactants, MSs are considered possible "green" alternatives to synthetic surfactants with better biodegradability, sustainability, and beneficial functional properties. This review therefore aims to explore the potential impacts of MSs as anti-aging ingredients.
Collapse
Affiliation(s)
- Amir Mohammad Bagheri
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Mirzahashemi
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Soodeh Salarpour
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasmin Dehghnnoudeh
- Departeman of Biology, Faculty of Science, York University, Toronto, Ontario, Canada
| | - Ibrahim M Banat
- School of Biomedical Sciences, Faculty of Life & Health Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - Mandana Ohadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Dehghannoudeh
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
9
|
Lu Y, Li T, Song L, Fan Q, Wang D, Wang P, Han Y, Zhou X. MDSCs in Chronic Liver Disease: Updates and Future Challenges. J Gastroenterol Hepatol 2025. [PMID: 40405825 DOI: 10.1111/jgh.17008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/14/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of pathologically expanded immature myeloid cells originating from bone marrow precursors, characterized by their potent immunosuppressive activity through mechanisms such as T cell inhibition, cytokine dysregulation, and metabolic interference. These cells are critically implicated in diverse pathological contexts, including cancer progression, chronic infections, and inflammatory disorders. In chronic liver diseases, MDSCs contribute to the pathogenesis of multiple conditions, such as chronic viral hepatitis, alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), and autoimmune liver diseases (AILD). Emerging evidence highlights their dual roles in both exacerbating tissue injury and modulating immune responses, positioning MDSCs as pivotal regulators of disease progression and potential therapeutic targets. In this review, we summarize the biological roles of MDSCs in a variety of chronic inflammatory liver diseases and explore the therapeutic potential of targeting these diseases to provide new insight for the treatment of chronic liver diseases.
Collapse
Affiliation(s)
- Yi Lu
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
- Department of Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Ting Li
- Department of Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Liang Song
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Qingling Fan
- Department of Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Danlin Wang
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Punan Wang
- Department of Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Ying Han
- Department of Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Xinmin Zhou
- Department of Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Liu Z, Chen J, Xu M, Ho S, Wei Y, Ho HP, Yong KT. Engineered multi-domain lipid nanoparticles for targeted delivery. Chem Soc Rev 2025. [PMID: 40390667 DOI: 10.1039/d4cs00891j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Engineered lipid nanoparticles (LNPs) represent a breakthrough in targeted drug delivery, enabling precise spatiotemporal control essential to treat complex diseases such as cancer and genetic disorders. However, the complexity of the delivery process-spanning diverse targeting strategies and biological barriers-poses significant challenges to optimizing their design. To address these, this review introduces a multi-domain framework that dissects LNPs into four domains: structure, surface, payload, and environment. Engineering challenges, functional mechanisms, and characterization strategies are analyzed across each domain, along with a discussion of advantages, limitations, and in vivo fate (e.g., biodistribution and clearance). The framework also facilitates comparisons with natural exosomes and exploration of alternative administration routes, such as intranasal and intraocular delivery. We highlight current characterization techniques, such as cryo-TEM and multiscale molecular dynamics simulations, as well as the recently emerging artificial intelligence (AI) applications-ranging from LNP structure screening to the prospective use of generative models for de novo design beyond traditional experimental and simulation paradigms. Finally, we examine how engineered LNPs integrate active, passive, endogenous, and stimuli-responsive targeting mechanisms to achieve programmable delivery, potentially surpassing biological sophistication in therapeutic performance.
Collapse
Affiliation(s)
- Zhaoyu Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Jingxun Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Mingkun Xu
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, China
| | - Sherwin Ho
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90095, USA.
| | - Yuanyuan Wei
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California, 90095, USA.
| | - Ho-Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, 999077, China.
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.
- The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
11
|
Yigci D, Ergönül Ö, Tasoglu S. Mpox diagnosis at POC. Trends Biotechnol 2025:S0167-7799(25)00160-X. [PMID: 40393854 DOI: 10.1016/j.tibtech.2025.04.015] [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: 11/17/2024] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/22/2025]
Abstract
The increasing number of Monkeypox (Mpox) cases in non-endemic countries resulted in the WHO declaring a public health emergency of international concern. Accurate and timely diagnosis of Mpox has a critical role in containing the spread of infection. Diagnosis currently relies on PCR, which requires trained personnel and complex laboratory infrastructure. Thus, the development of point-of-care (POC) tools are essential to facilitate rapid, accurate, and user-friendly diagnosis. Here, we review POC diagnostic tools available for Mpox. We also discuss bottlenecks preventing the widespread implementation of POC platforms for Mpox diagnosis and potential strategies to address these limitations. Furthermore, we describe future directions, including the role of machine learning (ML) and deep learning (DL)-based models and the integration of integrated field-deployable platforms for Mpox diagnosis.
Collapse
Affiliation(s)
- Defne Yigci
- School of Medicine, Koç University, Istanbul, 34450, Türkiye
| | - Önder Ergönül
- Koç University İşbank Center for Infectious Diseases, Istanbul, 34010, Türkiye; Department of Infectious Diseases and Clinical Microbiology, Koç University School of Medicine, Istanbul, 34010, Türkiye
| | - Savas Tasoglu
- Department of Mechanical Engineering, Koç University, Sariyer, Istanbul, 34450, Türkiye; Koç University Translational Medicine Research Center (KUTTAM), Koç University, Istanbul, 34450, Türkiye; Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Istanbul, 34684, Türkiye; Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Istanbul, 34450, Türkiye.
| |
Collapse
|
12
|
Li X, Sun W, Xu X, Jiang Q, Shi Y, Zhang H, Yu W, Shi B, Wan S, Liu J, Song W, Zhang J, Yuan Z, Li J. Hepatitis B virus surface antigen drives T cell immunity through non-canonical antigen presentation in mice. Nat Commun 2025; 16:4591. [PMID: 40382385 PMCID: PMC12085615 DOI: 10.1038/s41467-025-59985-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Hepatitis B virus (HBV) exclusively infects hepatocytes and produces large amounts of subviral particles containing its surface antigen (HBsAg). T cell immunity is crucial for controlling and clearing HBV infection. However, the intercellular processes underlying HBsAg presentation to T cells are incompletely understood. Here, using preclinical mouse models, we show that, following HBsAg expression, the intrahepatic Batf3+XCR1+CCR7- conventional dendritic cell subset cDC1 presents HBsAg by MHC-I cross-dressing, driving CD8+ T cell response. Meanwhile, upon HBsAg access to lymphoid tissues, B cells acquire HBsAg directly in the follicles of lymphoid tissues and initiate CD4+ T cell responses sequentially in the follicular and interfollicular regions, guided by chemoattractant receptors CCR5 and EBI2, respectively. Finally, we identify ALCAM, LFA-1, and CD80 as key co-stimulatory signals essential for optimal T cell responses. Thus, these findings reveal the roadmap of non-canonical antigen presentation that drives T cell immunity against HBsAg, advancing novel therapeutic strategies for chronic HBV infection.
Collapse
Affiliation(s)
- Xiaofang Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxuan Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaolan Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qirong Jiang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuheng Shi
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huixi Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weien Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Bisheng Shi
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Simin Wan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiangxia Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wuhui Song
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiming Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jianhua Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Isaia HA, Clerici NJ, Brandelli A. Bacillus lipopeptides as versatile antimicrobial weapons: looking toward antiviral activity. Crit Rev Biotechnol 2025:1-17. [PMID: 40368586 DOI: 10.1080/07388551.2025.2499152] [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: 02/13/2024] [Revised: 01/07/2025] [Accepted: 02/10/2025] [Indexed: 05/16/2025]
Abstract
Viral outbreaks are a topic of worldwide concern, resulting in a significant impact in health systems, a large number of deaths, and huge economical losses. The damage caused by Covid-19 has further highlighted the importance of prospecting for new molecules that can be applied in the prevention and treatment of viral infections. Many studies describe the remarkable antimicrobial activity of lipopeptides produced by Bacillus spp., especially against fungi and bacteria. However, research regarding the antagonistic effects on viruses is less frequent. Despite that, the antiviral activity of lipopeptides produced by Bacillus spp. has been demonstrated, indicating that these molecules could be potential candidates to control viral diseases. In this article, a compilation of reports with consistent data regarding the antiviral effect of Bacillus lipopeptides and the mechanisms involved in this process are presented. Moreover, the immunomodulatory role and toxicity profile of these molecules are discussed. Bacillus lipopeptides may exert an indirect antiviral effect, since they are able to positively induce humoral and cell-mediated immune responses. Moreover, their antiviral effect was observed in vitro and in vivo at nontoxic concentrations, offering a safe perspective for possible clinical application of these molecules. Finally, the challenges related to optimization and increasing production yield are addressed. This is the first critical review dedicated exclusively to antiviral activity of Bacillus lipopeptides.
Collapse
Affiliation(s)
- Henrique Ataide Isaia
- Laboratório de Nanobiotecnologia e Microbiologia Aplicada, Departamento de Ciência de Alimentos, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Naiara Jacinta Clerici
- Laboratório de Nanobiotecnologia e Microbiologia Aplicada, Departamento de Ciência de Alimentos, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adriano Brandelli
- Laboratório de Nanobiotecnologia e Microbiologia Aplicada, Departamento de Ciência de Alimentos, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
14
|
Yang P, Su W, Wang L, Xu F, Kong Y, Long J. From aldehyde metabolism to delay aging: targeting ALDH2 as a novel strategy. Free Radic Biol Med 2025; 236:70-86. [PMID: 40349798 DOI: 10.1016/j.freeradbiomed.2025.05.389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/19/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Aldehydes are molecules that are commonly found in both human physiology and the environment. The accumulation of these substances can lead to the cross-linking of intracellular DNA and proteins, thereby disrupting cellular function and contributing to the processes of premature aging and age-related diseases. Aldehyde dehydrogenase 2 (ALDH2), the key member of ALDH family, is an enzyme responsible for aldehyde metabolism, composed of four identical subunits located within the mitochondrial matrix. Its primary role is to catalyze the oxidation of aldehydes, resulting in the formation of their corresponding acid metabolites. This paper presents a succinct overview of the sources and metabolic pathways of key aldehydes within the human body, compares the various primary enzymes involved in aldehyde metabolism, and explores the structural and functional characteristics of ALDH2. Furthermore, ALDH2 is proposed as a potential therapeutic target for addressing aging and associated diseases. The discussion also includes prospective research avenues, particularly focusing on ALDH2 agonists and aldehyde scavengers designed to enhance the clearance of reactive aldehydes and safeguard cellular functions, thereby mitigating aldehyde-induced cellular damage and potentially delaying the aging process.
Collapse
Affiliation(s)
- Peng Yang
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Wu Su
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Lizhuo Wang
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Fanding Xu
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Yu Kong
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Jiangang Long
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China.
| |
Collapse
|
15
|
Kumari R, Banerjee S. Regulation of Different Types of Cell Death by Noncoding RNAs: Molecular Insights and Therapeutic Implications. ACS Pharmacol Transl Sci 2025; 8:1205-1226. [PMID: 40370994 PMCID: PMC12070317 DOI: 10.1021/acsptsci.4c00681] [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: 11/25/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Noncoding RNAs (ncRNAs) are crucial regulatory molecules in various biological processes, despite not coding for proteins. ncRNAs are further divided into long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) based on the size of their nucleotides. These ncRNAs play crucial roles in transcriptional, post-transcriptional, and epigenetic regulation. The regulatory roles of noncoding RNAs, including lncRNAs, miRNAs, and circRNAs, are essential in various modalities of cellular death, such as apoptosis, ferroptosis, cuproptosis, pyroptosis, disulfidptosis, and necroptosis. These noncoding RNAs are integral to modulating gene expression and protein functionality during cellular death mechanisms. In apoptosis, lncRNAs, miRNAs, and circRNAs influence the transcription of apoptotic genes. In ferroptosis, these noncoding RNAs target genes and proteins involved in iron homeostasis and oxidative stress responses. For cuproptosis, noncoding RNAs regulate pathways associated with the accumulation of copper ions, leading to cellular death. During pyroptosis, noncoding RNAs modulate inflammatory mediators and caspases, affecting the proinflammatory cell death pathway. In necroptosis, noncoding RNAs oversee the formation and functionality of necrosomes, thereby influencing the balance between cellular survival and death. Disulfidptosis is a unique type of regulated cell death caused by the excessive formation of disulfide bonds within cells, leading to cytoskeletal collapse and oxidative stress, especially under glucose-limited conditions. This investigation highlights the complex mechanisms through which noncoding RNAs coordinate cellular death, emphasizing their therapeutic promise as potential targets, particularly in the domain of cancer treatment.
Collapse
Affiliation(s)
- Reshmi Kumari
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Satarupa Banerjee
- Department of Biotechnology, School
of Biosciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
16
|
Wang Y, Feng X, Li Y, Niu S, Li J, Shi H, Wang G, Wang L. Targeting inflammation and necroptosis in diabetic kidney disease: A novel approach via PPARα modulation. Int Immunopharmacol 2025; 154:114562. [PMID: 40174339 DOI: 10.1016/j.intimp.2025.114562] [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: 01/24/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Renal tubular interstitial inflammation is a central driver of the pathogenesis of diabetic kidney disease (DKD). Peroxisome proliferator-activated receptor alpha (PPARα), predominantly expressed in renal tubular epithelial cells (TECs), plays a key role in regulating inflammation. However, the precise molecular mechanisms through which PPARα exerts its protective effects in DKD remain unclear. METHODS Single-cell RNA sequencing data from the GEO database revealed a marked reduction in PPARα expression in the proximal TECs of early-stage DKD patients. To investigate its potential role, we utilized an AAV9-PPARα viral vector to induce PPARα overexpression in TECs within a DKD mouse model. RNA sequencing of kidney tissues from both DKD and PPARα-overexpressing DKD mice was performed to identify key differentially expressed genes and signaling pathways. These findings were subsequently validated by in vitro and in vivo experiments. RESULTS PPARα overexpression significantly improved renal function, reduced interstitial fibrosis, attenuated inflammatory cytokine expression, and markedly decreased M1 macrophage infiltration. Notably, PPARα inhibited RIP1/RIP3/MLKL-mediated necroptosis in TECs, resulting in a substantial delay in DKD progression. Furthermore, NF-κB signaling played a crucial role in PPARα-mediated regulation of inflammation and necroptosis in TECs. CONCLUSION In summary, PPARα plays a pivotal role in modulating inflammation and necroptosis in DKD. Targeting PPARα in TECs represents a promising therapeutic strategy for slowing the progression of DKD and potentially reversing early renal damage. These findings open up new avenues for PPARα-targeted therapies in DKD and other chronic kidney diseases.
Collapse
Affiliation(s)
- Yu Wang
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Xiaojian Feng
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Yue Li
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Songlin Niu
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Jinxin Li
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Honghong Shi
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Gaoling Wang
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Lihua Wang
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
17
|
Abdelazim H, Barnes A, Stupin J, Hasson R, Muñoz-Ballester C, Young KL, Robel S, Smyth JW, Lamouille S, Chappell JC. Optimized enrichment of murine blood-brain barrier vessels with a critical focus on network hierarchy in post-collection analysis. Sci Rep 2025; 15:15778. [PMID: 40328881 PMCID: PMC12056178 DOI: 10.1038/s41598-025-99364-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Cerebrovascular networks contain a unique region of interconnected capillaries known as the blood-brain barrier (BBB). Positioned between upstream arteries and downstream veins, these microvessels have unique structural features, such as the absence of vascular smooth muscle cells (vSMCs) and a relatively thin basement membrane, to facilitate highly efficient yet selective exchange between the circulation and the brain interstitium. This vital role in neurological health and function has garnered significant attention from the scientific community and inspired methodology for enriching BBB capillaries. Extensive characterization of the isolates from such protocols is essential for framing the results of follow-on experiments and analyses, providing the most accurate interpretation and assignment of BBB properties. Seeking to aid in these efforts, here we visually screened output samples using fluorescent labels and found considerable reduction of non-vascular cells following density gradient centrifugation (DGC) and subsequent filtration. Comparatively, this protocol enriched brain capillaries, though larger diameter vessels associated with vSMCs could not be fully excluded. Protein analysis further underscored the enrichment of vascular markers following DGC, with filtration preserving BBB-associated markers and reducing - though not fully removing - arterial/venous contributions. Transcriptional profiling followed similar trends of DGC plus filtration generating isolates with less non-vascular and non-capillary material included. Considering vascular network hierarchy inspired a more comprehensive assessment of the material yielded from brain microvasculature isolation protocols. This approach is important for providing an accurate representation of the cerebrovascular segments being used for data collection and assigning BBB properties specifically to capillaries relative to other regions of the brain vasculature.
Collapse
Affiliation(s)
- Hanaa Abdelazim
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA
| | - Audra Barnes
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA
- Department of Biomedical Engineering and Mechanics and School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - James Stupin
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Ranah Hasson
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA
| | - Carmen Muñoz-Ballester
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, 21250, USA
| | - Kenneth L Young
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Stefanie Robel
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - James W Smyth
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA
- Department of Biomedical Engineering and Mechanics and School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24016, USA
| | - Samy Lamouille
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24016, USA
| | - John C Chappell
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), 2 Riverside Circle, Roanoke, VA, 24016, USA.
- FBRI Center for Vascular and Heart Research, Roanoke, VA, 24016, USA.
- Department of Biomedical Engineering and Mechanics and School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA.
| |
Collapse
|
18
|
Wan H, Yang Y, Tu Z, Tang M, Jing B, Feng Y, Xie J, Gao H, Song X, Zhao X. Enhanced mucosal immune response through nanoparticle delivery system based on chitosan-catechol and a recombinant antigen targeted towards M cells. Int J Biol Macromol 2025; 306:141345. [PMID: 40010449 DOI: 10.1016/j.ijbiomac.2025.141345] [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: 11/20/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
In mucosal vaccination, the targeted delivery of antigens through M (microfold) cells is essential for initiating a robust antigen-specific immune response. In the present study, we devised a nano-delivery platform to target M cells. This platform involved coating mesoporous silica nanoparticles (MSN) with a mucoadhesive chitosan-catechol (Chic) layer, incorporating a recombinant antigen to form nanoparticles that enhance the immune response. The collagenase equivalent domain (COE) of porcine epidemic diarrhea virus (PEDV) terminated with the M cell-targeting sequence RGD (COER), was initially expressed by Escherichia coli (E. coli) and subsequently conjugated to the surface of MSN-Chic, forming the MSN-Chic-COER nanoparticles. MSN-Chic-COER with strong mucoadhesive properties and a propensity for M cell targeting, demonstrated enhanced uptake by dendritic cells (DCs) and trafficking to lymph nodes, compared to COE/COER after intranasal administration. MSN-Chic-COER recruited more dendritic cells to the antigen-located site via stimulating chemokine CCL20 secretion was evidenced by cell co-culture model. Additionally, it enhanced antigen permeability by disrupting the distribution of the ZO-1 protein in epithelial cells. Notably, MSN-Chic-COER elicited a higher level of cellular immunity, humoral immunity, and PEDV neutralizing antibody production. These findings underscore the potential of MSN-Chic-COER as a promising intranasal vaccine delivery system.
Collapse
Affiliation(s)
- Hongping Wan
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yunhan Yang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiwen Tu
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingrun Tang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yumei Feng
- Instrumental analysis center, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiulong Xie
- Instrumental analysis center, Sichuan Agricultural University, Chengdu 611130, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China.
| | - Xu Song
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinghong Zhao
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
19
|
Morgan MJ, Kim YS. RIPK3 in necroptosis and cancer. Mol Cells 2025; 48:100199. [PMID: 40010643 PMCID: PMC11938148 DOI: 10.1016/j.mocell.2025.100199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Receptor-interacting protein kinase-3 is essential for the cell death pathway called necroptosis. Necroptosis is activated by the death receptor ligands and pattern recognition receptors of the innate immune system, leading to significant consequences in inflammation and in diseases, particularly cancer. Necroptosis is highly proinflammatory compared with other modes of cell death because cell membrane integrity is lost, resulting in releases of cytokines and damage-associated molecular patterns that potentiate inflammation and activate the immune system. We discuss various ways that necroptosis is triggered along with its potential role in cancer and therapy.
Collapse
Affiliation(s)
- Michael J Morgan
- Department of Natural Sciences, Northeastern State University, Tahlequah, OK 74464, USA.
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Ajou University, Suwon 16499, Korea; Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
20
|
Santos D, Carrijo Oliveira N, Costa ECA, Ramalho Paes MV, Beltrão-Braga B, Castanha AG, Beltrão-Braga PCB. Modeling potential drugs for Zika virus in animal and in vitro platforms: what is the current state of the art? Expert Opin Drug Discov 2025; 20:585-597. [PMID: 40251755 DOI: 10.1080/17460441.2025.2496461] [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: 02/06/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION The Zika virus (ZIKV) poses a significant public health threat due to its association with congenital Zika syndrome (CZS) and severe neurological disorders. Since its discovery, ZIKV has transitioned from sporadic outbreaks to a major epidemic in Brazil in 2015, which highlighted the urgent need for effective therapies, especially for vulnerable groups like pregnant women and newborns. AREAS COVERED This review provides a comprehensive overview of recent advancements in ZIKV drug discovery and their current stage of development, with a particular focus on those tested in animal models from 2018 to date, excluding vaccine candidates. Repurposed drugs, such as molnupiravir and sofosbuvir, have shown the potential to inhibit viral replication and mitigate disease. Novel compounds targeting viral proteins and host-directed therapies are also discussed. Furthermore, advanced in vitro models, including brain organoids, have offered critical insights into therapeutic efficacy. EXPERT OPINION Although some preclinical models have identified potential drugs ready for human translation, no protocol has yet been established for treating ZIKV infection. Currently, the treatment involves supportive care, managing symptoms, and preventing complications, especially for pregnant women. Ongoing research aims to develop specific antiviral therapies and vaccines; however, no such options are currently available.
Collapse
Affiliation(s)
- Debora Santos
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nathalia Carrijo Oliveira
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Institut Pasteur de São Paulo, São Paulo, Brazil
| | | | - Maria Vitória Ramalho Paes
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruna Beltrão-Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrelissa Gorete Castanha
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Institut Pasteur de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
21
|
Grycová A, Vyhlídalová B, Dvořák Z. The role of aryl hydrocarbon receptor in antiviral immunity: a focus on RNA viruses. Crit Rev Microbiol 2025:1-15. [PMID: 40299755 DOI: 10.1080/1040841x.2025.2497789] [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: 11/29/2024] [Revised: 02/02/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Aryl hydrocarbon receptor (AhR) is a ligand-dependent transcriptional factor that is activated by a plethora of exogenous and endogenous compounds, including environmental pollutants, drugs, and microbial metabolites. The AhR plays an important role in modulating immunity. Current findings suggest that AhR activation serves as a mechanism for evasion of host antiviral immune response and promotes viral replication. This review will focus on AhR's role in RNA virus infection because they show high mutation rates compared with DNA viruses, and therefo pose one of the greatest threats to humans in terms of potential pandemic risk. Indeed, they include human immunodeficiency virus (HIV), influenza A virus (IAV), coronaviruses (CoVs), Zika virus, and others. Understanding the mechanisms by which AhR influences the immune response to these viruses is critical for developing effective therapeutic strategies. By focusing on the interplay between AhR signaling and RNA virus infections, this review aims to contribute to the growing body of knowledge regarding host-pathogen interactions and the implications for antiviral immunity.
Collapse
Affiliation(s)
- Aneta Grycová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Barbora Vyhlídalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
22
|
Pavelescu LA, Mititelu-Zafiu NL, Mindru DE, Vladareanu R, Curici A. Molecular Insights into HPV-Driven Cervical Cancer: Oncoproteins, Immune Evasion, and Epigenetic Modifications. Microorganisms 2025; 13:1000. [PMID: 40431173 PMCID: PMC12113743 DOI: 10.3390/microorganisms13051000] [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: 03/28/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Cervical cancer ranks third in mortality and fourth in incidence among women worldwide as one of the leading causes of death from cancer in females. The main reason behind cervical carcinogenesis is long-term infection with high-risk human papillomavirus (HPV) genotypes, particularly HPV16 and HPV18. This review investigates HPV distribution across the world, along with cervical cancer molecular development mechanisms and current treatment strategies. Epidemiological data show that disease patterns vary significantly between different geographic regions because underdeveloped nations bear a higher disease burden. The molecular mechanisms of oncogenes E6 and E7 disrupt tumor suppressor pathways, while epigenetic modifications through DNA methylation and miRNA dysregulation promote malignant cell transformation. The reduction in HPV infection through prophylactic vaccination has shown promise, yet barriers related to accessibility and coverage still exist. The therapeutic technologies of gene expression inhibitors together with immunotherapies and epigenetic targeting agents show promise but require optimization to achieve specific targeting while minimizing off-target effects. A combined approach that integrates HPV vaccination with early diagnosis and molecular-specific therapies represents the most effective method to manage cervical cancer impact. The future care of patients will require increased translational research along with better immunization programs to drive prevention and therapeutic outcomes.
Collapse
Affiliation(s)
- Luciana Alexandra Pavelescu
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | | | - Dana Elena Mindru
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Radu Vladareanu
- Department of Obstetrics-Gynecology and Neonatology, Elias Emergency Hospital Bucharest, 011461 Bucharest, Romania
- Obstetrics and Gynecology, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania
| | - Antoanela Curici
- Department of Cellular and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Synevo Romania, 021408 Bucharest, Romania;
| |
Collapse
|
23
|
Lan X, Ma L, Ma J, Huang Z, Liu L, Li F, Wang M, Hu Y. Tas2r105 ameliorates gut inflammation, possibly through influencing the gut microbiota and metabolites. mSystems 2025; 10:e0155624. [PMID: 40079578 PMCID: PMC12013267 DOI: 10.1128/msystems.01556-24] [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: 11/20/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated gastrointestinal disorder that significantly impacts the life quality of people worldwide. Genetic factors play crucial roles in the development of IBD. Tas2rs, members of the G protein-coupled receptor (GPCR) superfamily, are known for their roles in bitter taste perception. However, Tas2rs have also been identified in the gut, where they help sense luminal contents and regulate gastrointestinal hormones. Periodontal Tas2r105 has been shown to modulate innate immunity by interacting with metabolites produced by oral bacteria. In this study, we observed increased Tas2r105 in the inflammatory colons induced by dextran sulfate sodium salt (DSS). We also noted that α-gustducin, the α-subunit of GPCRs, is present in the intestine, and that α-gustducin knockout mice exhibit aggravated colitis. Based on these findings, we hypothesize that Tas2r105 may play a role in immune regulation during IBD pathogenesis. To test this hypothesis, we used Tas2r105 knockout (KO) mice in a colitis model. Our results show that the KO mice had significantly shorter colon length, more severe colon inflammation, and greater destruction of the gut barrier compared with control mice. We also observed increased recruitment of macrophages to the lamina propria mucosa in the KO mice. Microbiological analysis revealed a significant increase in Proteobacteria and Bacteroidota, with a concomitant decrease in Firmicutes after Tas2r105 knockout. Metabolomic analysis showed a significant reduction in lysophosphatidylethanolamine (LPE) levels in the KO mice, which is known to have anti-inflammatory effects. Based on these findings, we speculate that Tas2r105 may help protect the intestine from inflammation by influencing the gut microbiota composition and LPE production.IMPORTANCEIncreased Tas2r105 was detected in the inflamed colon of mice outside the tongue. Tas2r105 deletion aggravated mice colon colitis. Tas2r105 might alleviate mice colitis by downregulating the Proteobacteria and the Bacteroidota abundance in the colon. Lysophosphatidylethanolamine (LPE) might be the key metabolite that mediated the intestinal protection of Tas2r105.
Collapse
Affiliation(s)
- Xiucai Lan
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liang Ma
- Department of Radiology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jiaming Ma
- Department of Health-Related Product Assessment, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Zhipeng Huang
- Departments of Gastroenterology, First Hospital of Quanzhou affiliated to Fujian Medical University, Quanzhou, China
| | - Lingling Liu
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Feng Li
- Department of Laboratory Animal Science, Shanghai Public Health Clinical Center, Shanghai, China
| | - Mingbang Wang
- Department of Neonatology, Affiliated Shenzhen Women and Children's Hospital (Longgang) of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, Guangdong, China
- Department of Experiment & Research, South China Hospital, Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yaomin Hu
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
24
|
Sun Z, Jiang W, Lu G, Ding Y, Wang L, Geng J, Zhang N, Wang H, Kang P, Tang B. Loss of ALDH2 accelerates the progression of pulmonary arterial hypertension through the 4-HNE/ERK1/2-p16 INK4a signaling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167863. [PMID: 40274079 DOI: 10.1016/j.bbadis.2025.167863] [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: 03/28/2024] [Revised: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Senescence is an important causative factor in the development of pulmonary arterial hypertension (PAH). Aldehyde dehydrogenase 2 (ALDH2), an enzyme involved in aldehyde detoxification, plays a role in cardiovascular diseases associated with aldehyde accumulation. This study aimed to investigate the role of ALDH2 in hypoxia-induced pulmonary arterial smooth muscle cells (PASMCs) and PAH. ALDH2 knockout (ALDH2-/-) mice and wild-type (WT) mice were exposed to a hypoxic environment with 10 ± 0.5 % oxygen concentration for 4 weeks to develop a chronic hypoxia-induced PAH (HPH) mouse model. We found that right ventricular hypertrophy and pulmonary arteriole muscularization were more severe in ALDH2-/- mice compared to WT mice. Additionally, ALDH2-/- mice exhibited elevated expression levels of 4-HNE, p-ERK1/2, the senescence-related protein p16INK4a, and the senescence-associated secretory phenotype (SASP) compared to WT mice. Similarly, treatment with the ALDH2 inhibitor (Daidzin) significantly increased 4-HNE, p-ERK1/2, p16INK4a, and SASP levels in PASMCs under hypoxia. Conversely, overexpression of ALDH2 reduced 4-HNE, p-ERK1/2, and PASMC senescence. Furthermore, exogenous 4-HNE, used to simulate hypoxia conditions, activated the ERK signaling pathway and induced PASMC senescence. However, ERK-specific inhibitors (PD98059) blocked hypoxia-induced PASMC senescence. These results demonstrate that ALDH2 deficiency induces PASMC senescence and promotes pulmonary vascular remodeling through the 4-HNE/ERK1/2-p16INK4a signaling pathway in HPH, providing a novel target for PAH treatment.
Collapse
Affiliation(s)
- Zhengyu Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Basic and Clinical Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui Province 233004, PR China
| | - Wendi Jiang
- Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui 233000, PR China; Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui 233000, PR China
| | - Guoqing Lu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Basic and Clinical Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui Province 233004, PR China
| | - Yangyang Ding
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Basic and Clinical Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui Province 233004, PR China
| | - Lei Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Basic and Clinical Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui Province 233004, PR China
| | - Jiayi Geng
- Department of Physiology, Bengbu Medical University, Bengbu, Anhui 233000, PR China
| | - Ningning Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Basic and Clinical Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui Province 233004, PR China
| | - Hongju Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China
| | - Pinfang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China; Key Laboratory of Basic and Clinical Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu, Anhui Province 233004, PR China.
| | - Bi Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, PR China.
| |
Collapse
|
25
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [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: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
26
|
Yu Z, Xu Z, Fu T, Liu S, Cui J, Zhang B, Liang J, Pang C, Ke Y, Wang R, Tang Z, Gao Y, Du B, Feng Y, Zhao H, Xue G, Yan C, Gan L, Feng J, Fan Z, Yang Y, Huang L, Zhao S, Ying S, Gu Q, Yuan J. Parallel comparison of T cell and B cell subpopulations of adenoid hypertrophy and tonsil hypertrophy of children. Nat Commun 2025; 16:3516. [PMID: 40229254 PMCID: PMC11997228 DOI: 10.1038/s41467-025-58094-w] [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: 05/09/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
The adenoids and tonsils are important immune organs of the nasopharynx that often become hypertrophic in childhood because of recurrent pathogen infection. However, the differences in the immune microenvironment of adenoid hypertrophy (AH) and tonsil hypertrophy (TH) are unclear. Here, we show the epidemiological characteristics and peripheral blood cell indices of 1209 pediatric patients (1-15 years old) diagnosed with AH, and find that AH is often accompanied by TH and characterized by specific changes in immune cell types. Single-cell RNA sequencing analysis show that 12 paired AH and TH samples contain large numbers of B, T cells and some exhausted effector memory CD4+ T cells. Compared with matched TH, AH have more naïve B cells and regulatory CD4+ T cells and less plasma B cells. Weaker antigen presentation and more significant immunosuppression are also observed in AH. In contrast, the number and cytotoxicity of cytotoxic CD8+ T cells decrease with AH grade. These findings will help our understanding of the immune response to nasopharyngeal infection.
Collapse
Affiliation(s)
- Zihui Yu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziying Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Tongtong Fu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shiyu Liu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Military supplies and energy quality supervision station of Bejing, Beijing, 100071, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Zhang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Jieqiong Liang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Chong Pang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ruikun Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Zhijie Tang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Yagang Gao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zheng Fan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yang Yang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lijuan Huang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shuo Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Sun Ying
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qinglong Gu
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China.
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
27
|
Wang L, Mu Q, Zhang W, Zheng W, Zhu X, Yu Y, Wang Y, Xu W, Lu Z, Han X. Placental targeted drug delivery: a review of recent progress. NANOSCALE 2025; 17:8316-8335. [PMID: 40070242 DOI: 10.1039/d4nr05338a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
The placenta plays a crucial role in mediating nutrient and gas exchange between the mother and fetus during pregnancy. Targeting therapeutic agents to the placenta presents significant opportunities for treating placental disorders and enhancing fetal outcomes. However, the unique structural complexity and selective permeability of the placenta pose substantial challenges for effective drug delivery. This review provides a comprehensive overview of current strategies for placental targeting, including lipid nanoparticle (LNP) delivery systems, targeted peptide modifications, specific antibody targeting of placental receptors, and the use of viral vectors. We critically analyze the advantages and limitations of each approach, emphasizing recent advancements in enhancing targeting specificity and delivery efficiency. By consolidating the latest research developments, this review aims to foster further innovation in placental drug delivery methods and contribute significantly to the advancement of therapeutic strategies for placental disorders, ultimately improving outcomes for both mother and fetus.
Collapse
Affiliation(s)
- Linjian Wang
- Department of Obstetrics, Haining Maternal and Child Health Hospital, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No. 309, East Shuiyueting Road, Xiashi Street, Haining, Zhejiang, 314400, China
| | - Qiuqiu Mu
- Third Affliated Hospital of Wenzhou Medical University, WanSong Road No. 108, Ruian, Wenzhou, Zhejiang, 325200, China
| | - Wenjing Zhang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Weiqian Zheng
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Ying Yu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - YuPeng Wang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Wenli Xu
- Department of Obstetrics, Haining Maternal and Child Health Hospital, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No. 309, East Shuiyueting Road, Xiashi Street, Haining, Zhejiang, 314400, China
| | - Zhimin Lu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Xiujun Han
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
28
|
Zhang B, Fu T, Han Y, Li G, Wan X, Li Y. Experimental study of a novel mouse model of tibial shaft fracture combined with blunt chest trauma. Animal Model Exp Med 2025; 8:696-706. [PMID: 38225728 PMCID: PMC12008451 DOI: 10.1002/ame2.12379] [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: 07/05/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUD Thoracic Trauma and Limb Fractures Are the Two most Common Injuries in Multiple Trauma. However, there Is Still a Lack of Mouse Models of Trauma Combining Tibial Shaft Fracture (TSF) and Thoracic Trauma. In this Study, we Attempted to Develop a Novel Mouse Model of TSF Combined with Blunt Chest Trauma (BCT). METHODS A total of 84 C57BL/6J male mice were used as the multiple trauma model. BCT was induced by hitting the chests of mice with heavy objects, and TSF was induced by hitting the tibia of mice with heavy objects after intramedullary fixation. Serum specimens of mice were received by cardiac puncture at defined time points of 0, 6, 12, 24, 48, and 72 h. RESULTS Body weight and body temperature tended to decrease within 24 h after multiple trauma. Hemoglobin analyses revealed a decrease during the first 24 h after multiple trauma. Some animals died by cardiac puncture immediately after chest trauma. These animals exhibited the most severe pulmonary contusion and hemorrhage. The level of lung damage varied in diverse mice but was apparent in all animals. Classic hematoxylin and eosin (H&E)-stained paraffin pulmonary sections of mice with multiple trauma displayed hemorrhage and an immunoinflammatory reaction. Bronchoalveolar lavage fluid (BALF) and serum samples of mice with multiple trauma showed an upregulation of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-1α (TNF-1α) compared with the control group. Microimaging confirmed the presence of a tibia fracture and pulmonary contusion. CONCLUSIONS The novel mouse multiple trauma model established in this study is a common trauma model that shows similar pathological mechanisms and imaging characteristics in patients with multiple injuries. This study is useful for determining whether blockade or intervention of the cytokine response is beneficial for the treatment of patients with multiple trauma. Further research is needed in the future.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Emergency Intensive Care MedicineAffiliated Hospital of Yangzhou UniversityYangzhouChina
| | - Tingting Fu
- Department of Special Care Medicine, Affiliated Hospital of Yangzhou UniversityYangzhouChina
| | - Yi Han
- Department of Clinical MedicineMedical College of Yangzhou UniversityYangzhouChina
| | - Gongke Li
- Department of Emergency Intensive Care MedicineAffiliated Hospital of Yangzhou UniversityYangzhouChina
| | - Xianyao Wan
- Department of Critical Care MedicineAffiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yong Li
- Department of Intensive Care MedicineAffiliated Hospital of Yangzhou UniversityYangzhouChina
| |
Collapse
|
29
|
Wang C, Zai W, Zhao K, Li Y, Shi B, Wu M, Zhou X, Kozlowski M, Zhang X, Fang Z, Yuan Z. Potential role of liver-resident CD3 + macrophages in HBV clearance in a mouse hepatitis B model. JHEP Rep 2025; 7:101323. [PMID: 40143948 PMCID: PMC11937660 DOI: 10.1016/j.jhepr.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 03/28/2025] Open
Abstract
Background & Aims Chronic HBV infection usually causes cirrhosis and hepatocellular carcinoma. Comparative investigations of acute and chronic HBV cases would help determine the immune responses crucial for viral clearance. Methods A fast-cleared HBV mouse model was established in Alb-Cre mice via hydrodynamic injection of HBV plasmid, while persistent HBV model mice were generated via recombinant covalently closed circular DNA-adeno-associated virus 8 infection. The single-cell transcriptomes of CD45+ intrahepatic non-parenchymal cells from these mice were conducted. Multiplexed immunohistochemistry and flow cytometry were used to confirm the findings from single-cell transcriptomes. Transwell, coculture, and adoptive transfer experiments were performed to study the generation and functions of macrophages. Results Twenty-four clusters of immune cells were identified. Myeloid cells, including granulocytes, monocytes, and dendritic cells, are activated early in HBV fast-cleared mice. Significantly, a cluster of CD3+ macrophages was found in the viral clearance phase, which was confirmed in liver tissue from five acute patients with HBV. These cells highly expressed CXCL1, tumor necrosis factor alpha, and HBsAg-specific T cell receptors. The transwell assay revealed that CD3+ macrophages originate from macrophages (n = 6). T cells and anti-HBsAg antibodies are indispensable for their differentiation, which was further confirmed in T- and/or B-cell-deficient mice. Interestingly, these CD3+ macrophages capable of killing peptide-loaded hepatocytes and engulfing IgG-coated beads were persistently detectable in the mouse liver for 10 weeks after HBV clearance. The expression levels of CD5L and Bcl2, two classical antiapoptotic proteins, increased (p <0.001), suggesting that the CD3+ macrophages are long-term resident populations. Finally, adoptive transfer of CD3+ macrophages accelerated HBV clearance in mice (n = 5, p <0.01). Conclusions We identified long-term polyfunctional CD3+ macrophages residing in HBV fast-cleared livers that could help elucidate the immune responses involved in eliminating HBV. Impact and implications The liver is a special organ with unique immune characteristics and tolerance to foodborne antigens. Chronic infections can develop in newborns after exposure to HBV; however, acute infections usually occur in adults, indicating that immune cells in the liver tissue microenvironment can also effectively fight against the virus. Nevertheless, the mechanisms involved in acute HBV infection have rarely been studied. In this study, we identified a macrophage population with both T cell and macrophage characteristics in the livers of acute HBV model mice and revealed that these macrophages play important roles in HBV clearance. Moreover, we confirmed that this population is derived from macrophages in the presence of virus-specific T cells and antibodies. This finding highlights the complexity of antiviral immune responses in liver microenvironments.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kuangjie Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bisheng Shi
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Wu
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Maya Kozlowski
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaonan Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhong Fang
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Cavuto ML, Malpartida-Cardenas K, Pennisi I, Pond MJ, Mirza S, Moser N, Comer M, Stokes I, Eke L, Lant S, Szostak-Lipowicz KM, Miglietta L, Stringer OW, Mantikas KT, Sumner RP, Bolt F, Sriskandan S, Holmes A, Georgiou P, Ulaeto DO, Maluquer de Motes C, Rodriguez-Manzano J. Portable molecular diagnostic platform for rapid point-of-care detection of mpox and other diseases. Nat Commun 2025; 16:2875. [PMID: 40128193 PMCID: PMC11933461 DOI: 10.1038/s41467-025-57647-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
The World Health Organization's designation of mpox as a public health emergency of international concern in August 2024 underscores the urgent need for effective diagnostic solutions to combat this escalating threat. The rapid global spread of clade II mpox, coupled with the sustained human-to-human transmission of the more virulent clade I mpox in the Democratic Republic of Congo, highlights a critical gap in point-of-care diagnostics for this emergent disease. In response, we developed Dragonfly, a portable molecular diagnostic platform for point-of-care use that integrates power-free nucleic acid extraction (<5 minutes) with lyophilised colourimetric LAMP chemistry. The platform demonstrated an analytical limit-of-detection of 100 genome copies per reaction for monkeypox virus, effectively distinguishing it from other orthopoxviruses, herpes simplex virus, and varicella-zoster virus. Clinical validation on 164 samples, including 51 mpox-positive cases, yielded 96.1% sensitivity and 100% specificity for orthopoxviruses, and 94.1% sensitivity and 100% specificity for monkeypox virus. Here, we present a rapid, accessible, and robust point-of-care diagnostic solution for mpox, suitable for both low- and high-resource settings, addressing the global resurgence of orthopoxviruses in the context of declining smallpox immunity.
Collapse
Affiliation(s)
- Matthew L Cavuto
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Kenny Malpartida-Cardenas
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Ivana Pennisi
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Marcus J Pond
- Department of Infection and Immunity, Imperial College Healthcare NHS Trust, London, UK
| | - Sohail Mirza
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Nicolas Moser
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
- Department of Electrical and Electronic Engineering, Imperial College London, London, UK
| | - Mark Comer
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Isobel Stokes
- Department of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Lucy Eke
- Department of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Sian Lant
- Department of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | | | - Luca Miglietta
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Oliver W Stringer
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Katerina-Theresa Mantikas
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
- Department of Electrical and Electronic Engineering, Imperial College London, London, UK
| | - Rebecca P Sumner
- Department of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Frances Bolt
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
| | - Shiranee Sriskandan
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Alison Holmes
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- David Price Evans Infectious Diseases & Global Health Group, University of Liverpool, Liverpool, UK
- The Fleming Initiative, Imperial College London and Imperial College Healthcare NHS Trust, London, UK
| | - Pantelis Georgiou
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK
- Department of Electrical and Electronic Engineering, Imperial College London, London, UK
| | - David O Ulaeto
- CBR Division, Defence Science and Technology Laboratory, Salisbury, UK
| | - Carlos Maluquer de Motes
- Department of Microbial Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Jesus Rodriguez-Manzano
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK.
- ProtonDx Ltd, Translation & Innovation Hub, Imperial College London, London, UK.
| |
Collapse
|
31
|
Li P, Fan Z, Huang Y, Luo L, Wu X. Mitochondrial dynamics at the intersection of macrophage polarization and metabolism. Front Immunol 2025; 16:1520814. [PMID: 40196123 PMCID: PMC11973336 DOI: 10.3389/fimmu.2025.1520814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Macrophages are vital sentinels in innate immunity, and their functions cannot be performed without internal metabolic reprogramming. Mitochondrial dynamics, especially mitochondrial fusion and fission, contributes to the maintenance of mitochondrial homeostasis. The link between mitochondrial dynamics and macrophages in the past has focused on the immune function of macrophages. We innovatively summarize and propose a link between mitochondrial dynamics and macrophage metabolism. Among them, fusion-related FAM73b, MTCH2, SLP-2 (Stomatin-like protein 2), and mtSIRT, and fission-related Fis1 and MTP18 may be the link between mitochondrial dynamics and macrophage metabolism association. Furthermore, post-translational modifications (PTMs) of mtSIRT play prominent roles in mitochondrial dynamics-macrophage metabolism connection, such as deacetylates and hypersuccinylation. MicroRNAs such as miR-150, miR-15b, and miR-125b are also possible entry points. The metabolic reprogramming of macrophages through the regulation of mitochondrial dynamics helps improve their adaptability and resistance to adverse environments and provides therapeutic possibilities for various diseases.
Collapse
Affiliation(s)
- Pan Li
- Department of Environment and Safety Engineering, Taiyuan Institute of Technology, Taiyuan, China
| | - Zhengbo Fan
- People’s Government of Huangshui Town, Shizhu Tujia Autonomous County, Chongqing, China
| | - Yanlan Huang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Liang Luo
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaoyan Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
32
|
Liu J, Ma S, Deng D, Yang Y, Li J, Zhang Y, Yin P, Shang D. Multi-Omics Profiling Reveals Glycerolipid Metabolism-Associated Molecular Subtypes and Identifies ALDH2 as a Prognostic Biomarker in Pancreatic Cancer. Metabolites 2025; 15:207. [PMID: 40137171 PMCID: PMC11943634 DOI: 10.3390/metabo15030207] [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: 02/26/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background: The reprogramming of lipid metabolism, especially glycerolipid metabolism (GLM), plays a key role in cancer progression and response to therapy. However, the role and molecular characterization of GLM in pancreatic cancer (PC) remain unclear. Methods: A pan-cancer analysis of glycerolipid metabolism-related genes (GMRGs) was first conducted to assess copy-number variants, single-nucleotide variations, methylation, and mRNA expression. Subsequently, GLM in PC was characterized using lipidomics, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomic analysis. A cluster analysis based on bulk RNA sequencing data from 930 PC samples identified GLM-associated subtypes, which were then analyzed for differences in prognosis, biological function, immune microenvironment, and drug sensitivity. To prioritize prognostically relevant GMRGs in PC, we employed a random forest (RF) algorithm to rank their importance across 930 PC samples. Finally, the key biomarker of PC was validated using PCR and immunohistochemistry. Results: Pan-cancer analysis identified molecular features of GMRGs in cancers, while scRNA-seq, spatial transcriptomics, and lipidomics highlighted GLM heterogeneity in PC. Two GLM-associated subtypes with significant prognostic, biofunctional, immune microenvironmental, and drug sensitivity differences were identified in 930 PC samples. Finally, ALDH2 was identified as a novel prognostic biomarker in PC and validated in a large number of datasets and clinical samples. Conclusions: This study highlights the crucial role of GLM in PC and defines a new PC subtype and prognostic biomarker. These findings establish a novel avenue for studying prognostic prediction and precision medicine in PC patients.
Collapse
Affiliation(s)
- Jifeng Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
| | - Dawei Deng
- Department of Hepato-Biliary-Pancreas, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China;
| | - Yao Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Junchen Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Yunshu Zhang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Peiyuan Yin
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
33
|
Kökrek E, Pir P. Distinct deregulation trends of transcriptional protein complexes in aging naive T cells. J Leukoc Biol 2025; 117:qiae231. [PMID: 39437255 DOI: 10.1093/jleuko/qiae231] [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: 08/02/2024] [Revised: 09/19/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024] Open
Abstract
The impact of aging on T cell subsets, specifically CD4+ and CD8+ T cells, leading to immune system dysfunction has been the focus of scientific investigation due to its potential to reverse age-associated deterioration. Transcriptomic and epigenomic studies have identified the primary regulators in T cell aging. However, comprehending the underlying dynamic mechanisms requires studying these proteins with their interactors. Here, we integrated single-cell RNA sequencing data of naive CD4+ and CD8+ T cells obtained from 3 different age groups with protein-protein and domain-domain interaction networks to predict and compare the transcriptional protein complexes and identify their capacity to explain age-associated variances. Our novel approach revealed significant effects of aging on the repertoire of complexes, which remains unchanged in naive CD4+ T cells, while in naive CD8+ T cells, it diminishes. In both cell types, there was major deregulation of complexes with the same composition, involving a range of transcription factors. This aging-associated deregulation is characterized by a specific set of protein complexes in naive CD4+ T cells, but this pattern is not observed in naive CD8+ T cells. SMAD3 and BCL11A complexes emerge as key markers in defining a trajectory in aging naive CD4+ T cells. These complexes can accurately distinguish between 3 different age groups, indicating their potential as targets. The direct link between SMAD3 and FOS complexes whose regulatory role has been previously implicated in aging and MBD3 as the novel key link between SMAD3 and BCL11A complexes implicates a coordinated mechanism in age-associated deregulation.
Collapse
Affiliation(s)
- Emel Kökrek
- Department of Molecular Biology and Genetics, Kadir Has University, Cibali, Kadir Has Cd., 34083 Fatih/Istanbul, Turkey
- Department of Bioengineering, Gebze Technical University, Cumhuriyet, 2254. Sk. No:2, 41400 Gebze/Kocaeli, Turkey
| | - Pınar Pir
- Department of Bioengineering, Gebze Technical University, Cumhuriyet, 2254. Sk. No:2, 41400 Gebze/Kocaeli, Turkey
| |
Collapse
|
34
|
Frigard R, Ajayi OM, LeFevre G, Ezemuoka LC, English S, Benoit JB. Daily activity rhythms, sleep, and pregnancy are fundamentally related in the Pacific beetle mimic cockroach, Diploptera punctata. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640076. [PMID: 40060505 PMCID: PMC11888445 DOI: 10.1101/2025.02.27.640076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Sleep and pregnancy are contentious bedfellows; sleep disorders and disturbances are associated with adverse pregnancy outcomes, although much is still unknown about this relationship. Sleep and pregnancy have been studied in many models, but most focus heavily on mammals. However, pregnancy is ubiquitous across the animal kingdom - a hallmark of convergent evolution; similarly sleep is a shared feature across diverse species. Here, we present an ideal model in which to study the dynamics between sleep and pregnancy in invertebrates. The Pacific beetle mimic cockroach, Diploptera punctata, is a viviparous cockroach species that uses milk proteins to nourish its young with a broodsac over a three month pregnancy. However, little is known about the relationship between this unique reproductive biology and daily rhythms of activity and sleep. We established that D. punctata displayed a peak in activity shortly following sunset, with males significantly more active than females. When scavenging behavior was examined, males and non-pregnant females emerged more often and traveled further from a shelter compared to pregnant females, suggesting reduced risk-taking behavior in late pregnancy. Chronic disturbance of sleep during pregnancy negatively impacted embryo development by increasing gestational duration and decreasing the transcription of milk proteins. These findings indicate that sleep is key to embryo development and that pregnancy has a significant impact on the daily rhythms of activity in Diploptera punctata. More broadly, we present a tractable invertebrate model for understanding the relationship between sleep and pregnancy, which will aid in the exploration of the poorly understood interface between these two ubiquitous and highly conserved traits.
Collapse
Affiliation(s)
- Ronja Frigard
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221
| | - Oluwaseun M Ajayi
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221
| | - Gabrielle LeFevre
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221
| | - Lilian C Ezemuoka
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221
| | - Sinead English
- School of Biological Sciences, University of Bristol, UK
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221
| |
Collapse
|
35
|
Xing L, Liu Z, Wang X, Liu Q, Xu W, Mao Q, Zhang X, Hao A, Xia S, Liu Z, Sun L, Zhang G, Wang Q, Chen Z, Jiang S, Sun L, Lu L. Early fusion intermediate of ACE2-using coronavirus spike acting as an antiviral target. Cell 2025; 188:1297-1314.e24. [PMID: 39889696 DOI: 10.1016/j.cell.2025.01.012] [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: 04/30/2024] [Revised: 08/28/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Coronavirus fusion with and entry into the host cell depends on viral spike, which acts as a crucial component of viral infection. However, the lack of receptor-activated spike intermediate conformation has hindered a comprehensive understanding of spike-induced membrane fusion. Here, we captured an angiotensin-converting enzyme 2 (ACE2)-induced early fusion intermediate conformation (E-FIC) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in which heptad repeat 1 (HR1) in S2 has ejected while S1 remains attached. This E-FIC can transition to the late FIC after S2' cleavage. Leveraging this discovery, we designed an E-FIC-targeted dual-functional antiviral protein, AL5E. AL5E effectively inactivated ACE2-using coronaviruses and inhibited their infection, outperforming a mono-functional antiviral in protecting animals against these coronaviruses. This study has identified the E-FIC and used it as a target for the development of a dual-functional antiviral for the prevention and treatment of ACE2-using coronavirus infection.
Collapse
Affiliation(s)
- Lixiao Xing
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Zhimin Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Xinling Wang
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Qianying Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Wei Xu
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Qiyu Mao
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Xiang Zhang
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Aihua Hao
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Shuai Xia
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Zezhong Liu
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Lujia Sun
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Guangxu Zhang
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Qian Wang
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Zhenguo Chen
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China.
| | - Lei Sun
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China.
| | - Lu Lu
- Shanghai Institute of Infectious Disease and Biosecurity, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Fudan University, Shanghai 200032, China.
| |
Collapse
|
36
|
Tang Z, Chen C, Zhou C, Liu Z, Li T, Zhang Y, Feng Y, Gu C, Li S, Chen J. Insights into tumor-derived exosome inhibition in cancer therapy. Eur J Med Chem 2025; 285:117278. [PMID: 39823808 DOI: 10.1016/j.ejmech.2025.117278] [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: 10/02/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Exosomes are critical mediators of cell-to-cell communication in physiological and pathological processes, due to their ability to deliver a variety of bioactive molecules. Tumor-derived exosomes (TDEs), in particular, carry carcinogenic molecules that contribute to tumor progression, metastasis, immune escape, and drug resistance. Thus, TDE inhibition has emerged as a promising strategy to combat cancer. In this review, we discuss the key mechanisms of TDE biogenesis and secretion, emphasizing their implications in tumorigenesis and cancer progression. Moreover, we provide an overview of small-molecule TDE inhibitors that target specific biogenesis and/or secretion pathways, highlighting their potential use in cancer treatment. Lastly, we present the existing obstacles and propose corresponding remedies for the future development of TDE inhibitors.
Collapse
Affiliation(s)
- Ziwei Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Zhouyan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Zhang
- School of Petrochemical Engineering, Changzhou University, Changzhou, 213164, China.
| | - Yanyan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chenglei Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijia Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
37
|
Gao Y, Zhang Y, Mi N, Miao W, Zhang J, Liu Y, Li Z, Song J, Li X, Guan W, Bai C. Exploring the link between M1 macrophages and EMT of amniotic epithelial cells: implications for premature rupture of membranes. J Nanobiotechnology 2025; 23:163. [PMID: 40033278 PMCID: PMC11877754 DOI: 10.1186/s12951-025-03192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/01/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Despite increasing evidence supporting the role of an amniotic epithelial-mesenchymal transition (EMT) in the premature rupture of membranes (PROMs), it remains unclear if extracellular vesicle (EV) derived from M1 macrophages play a critical role in triggering the EMT of amniotic epithelial cells (AECs). RESULTS This study revealed that under inflammatory conditions, EV-miR-146a/155 from M1 macrophages could trigger EMTs and MMP-9 transcription in AECs, elevating the risk of PROM in both mice and humans. Introduction of EV-miR-155 led to inhibition of Ehf expression and reduced E-cadherin transcription in AECs. Meanwhile, EV-miR-146a activated the β-catenin/Tcf7 complex to promote the transcription of Snail, MMP-9, and miR-146a/155, inducing EMTs. Subsequently, EMT induction in AECs is associated with a loss of epithelial characteristics, disruption of cellular junctions, widening of intercellular spaces, and diminished biomechanical properties of the amniotic membrane. CONCLUSION Inflammatory stimulation prompts the polarization of macrophages in amniotic fluid into the M1 type, which subsequently secrete EVs laden with inflammatory miRNAs. These EVs trigger the EMT of AECs, causing the loss of their epithelial phenotype. Consequently, the biomechanical properties of the amnion deteriorate, ultimately leading to its rupture, posing risks relevant to pregnancy complications such as premature rupture of membranes. The results of this study provide insights into the pathogenesis of PROM and will aid in treatment development.
Collapse
Affiliation(s)
- Yuhua Gao
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No.2 Yuanmingyuan West Street, Haidian District, Beijing, 100193, P. R. China
| | - Yanan Zhang
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, Jining, 272000, P. R. China
| | - Ningning Mi
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, No. 666, Wusu Road, Lin'an, 311300, P. R. China
| | - Wang Miao
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Jingmiao Zhang
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Yize Liu
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Zhikun Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Jiaxun Song
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Xiangchen Li
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, No. 666, Wusu Road, Lin'an, 311300, P. R. China.
| | - Weijun Guan
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No.2 Yuanmingyuan West Street, Haidian District, Beijing, 100193, P. R. China.
| | - Chunyu Bai
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China.
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No.2 Yuanmingyuan West Street, Haidian District, Beijing, 100193, P. R. China.
| |
Collapse
|
38
|
Liu M, Zhu Y, Meng W, Zhang C, Chen Y, Shi Q, Song S, Zheng S, Liu Y, Zhou Y, Chen G. Potential therapeutic effect of dimethyl fumarate on Treg/Th17 cell imbalance in biliary atresia. Clin Immunol 2025; 272:110439. [PMID: 39863040 DOI: 10.1016/j.clim.2025.110439] [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: 11/03/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
The imbalance between Tregs and proinflammatory Th17 cells in children with biliary atresia (BA) causes immune damage to cholangiocytes. Dimethyl fumarate (DMF), an immunomodulatory drug, regulates the Treg/Th17 balance in diseases like multiple sclerosis (MS). This study explores DMF's effect on Treg/Th17 balance in BA and its potential mechanism. The differential gene expression profiles in liver of BA and choledochal cyst (CC) patients were analyzed by single-cell RNA sequencing (scRNA-seq). Treg and Th17 cell frequencies in BA hilar lymph nodes (LNs) were determined by flow cytometry. CD3+ T cells were isolated from BA hilar LNs and treated with DMF in vitro to observe their differentiation. The effects of DMF were evaluated on BA mouse model, and enzyme-linked immunosorbent assay to measure biochemical markers and cytokine profiles. The Treg/Th17 ratio in the liver was determined by flow cytometry. Nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant genes solute carrier family 7 member 1 (Slc7a11), heme oxygenase - 1 (Hmox1) was validated by q-PCR and Western blot. ScRNA-seq showed CD4+ T cells in BA liver were enriched in antioxidant pathways. The Treg/Th17 ratio in BA hilar LNs was significantly reduced compared to CC. In vitro, DMF promoted Treg differentiation and inhibited Th17 differentiation. In vivo, the Treg/Th17 ratio increased in the liver of the DMF 40 mg/kg group. In the 40 mg/kg DMF group, interleukin-17 A (IL-17 A) expression decreased as seen in pathological staining. DMF increased Nrf2, Hmox1, Slc7a11 mRNA and protein levels in DMF 40 mg/kg group. There is a Treg/Th17 imbalance in BA patients' hilar LNs, which DMF can restore in vitro. DMF improves the survival rate of BA mice and corrects the Treg/Th17 imbalance, possibly via the Nrf2/antioxidant response elements (ARE) pathway.
Collapse
Affiliation(s)
- Mengting Liu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, China
| | - Ye Zhu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, China
| | - Weida Meng
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Caiyan Zhang
- Department of Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Yuke Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, China
| | - Qi Shi
- Department of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sun Song
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, China
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yufeng Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, 201102, China.
| | - Gong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, China.
| |
Collapse
|
39
|
Wang Z, Yang X, Wang ZZ, Yu XF, Li Z, Song S, Zhao Y, Kuang YQ, Li YY, Zhang C. A rapid and sensitive extraction-free HiFi-LAMP assay for detecting Mycobacterium leprae. Int J Infect Dis 2025; 152:107835. [PMID: 39929318 DOI: 10.1016/j.ijid.2025.107835] [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: 11/26/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Timely and accurate detection of Mycobacterium leprae (M. leprae) is crucial for efficient treatment and early intervention of leprosy, which requires a simple and rapid extraction-free assay. METHODS A HiFi-loop-mediated isothermal amplification (LAMP) assay was developed for detection of M. leprae. The performance of the assay was assessed by comparing with qPCR and nested PCR assays using clinical samples. The extraction-free HiFi-LAMP assay was assessed by saliva from individuals with leprosy. RESULTS The M. leprae HiFi-LAMP assay has high specificity and sensitivity with a limit of detection (LOD) of 43 copies/25 µL reaction. Both sensitivity and specificity of the HiFi-LAMP assay were 100% for 130 purified DNA from nasal and oral samples, and the sensitivity was slightly higher than 50%-88.9% by the qPCR assay. A higher detection rate of M. leprae was observed in nasal swabs than oral swabs. The extraction-free assay directly using 6 µL saliva has a LOD of 11,833 M. leprae RLEP copies/mL saliva, can be completed within 30 mins, and showed 66.7% sensitivity for three saliva samples when compared with the assay using purified DNA. CONCLUSION The standard and/or extraction-free HiFi-LAMP assays can be used for detecting and monitoring M. leprae in endemic areas in resource-limited settings.
Collapse
Affiliation(s)
- Zhengfang Wang
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China; Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xi Yang
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhi-Ze Wang
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Xiu-Feng Yu
- Wenshan Institute of Dermatology, Wenshan, China
| | - Zhe Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shangwen Song
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China; Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yongjuan Zhao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yi-Qun Kuang
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu-Ye Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Chiyu Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
40
|
Li J, Ma X, Xuan Q, Li Q, Wu M, Shi B, Fang Z, Chen L, Chen J, Wen Y, Zhu C, Zhu L, Zhang X, Yuan Z. Modulation of monocyte activity by hepatocellular MicroRNA delivery through HBsAg particles: Implications for pathobiology of chronic hepatitis B. Hepatology 2025; 81:990-1005. [PMID: 38904485 PMCID: PMC11825484 DOI: 10.1097/hep.0000000000000972] [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] [Received: 11/28/2023] [Accepted: 05/21/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND AND AIMS HBsAg serves as an important immune-modulatory factor in chronic hepatitis B. One aspect of such modulation may act through monocytes, which are the major Ag-presenting cells taking up HBsAg. There is evidence for the encapsulation of hepatocellular microRNAs (miRNAs) by HBsAg particles, while its pathobiological significance is unclear. Here, we characterized the miRNA profile in patients with chronic hepatitis B and probed their association with liver inflammation. APPROACHES AND RESULTS We collected plasma from patients that are treatment-naive with chronic hepatitis B (n = 110) and quantified total/HBsAg-enveloped miRNAs by qRT-PCR and plasma cytokines by ELISA. The biological effects of HBsAg-delivered miRNAs in monocytes were evaluated using multiple approaches. The clinical significance of candidate miRNAs and cytokines was corroborated in patients with HBV-associated advanced liver diseases. The plasma miRNA profile showed 2 major clusters, one significantly associated with HBsAg titer and the other correlated with liver inflammation. Among HBsAg-carried miRNAs, miR-939 displayed the most significant correlation with IL-8. Mechanistically, miR-939 in subviral particles enters monocytes and significantly augments IL-8 production through the mitogen-activated protein kinase (MAPK) p38 signaling pathway. Finally, the findings that miR-939 positively correlated with IL-8 level and inflammation/fibrosis stage in the cohort of HBV-associated advanced liver diseases support its causative role in the progression of liver diseases. CONCLUSIONS HBsAg particles carry hepatocellular miRNAs, including miR-939, which enter monocytes and alter their functional status, such as IL-8 secretion. Our findings demonstrate that the HBsAg-miR-939-IL-8 axis may play a crucial role in HBV-induced hepatic necro-inflammation and the progression of advanced liver diseases.
Collapse
Affiliation(s)
- Jin Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, China
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao Ma
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, China
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qinkao Xuan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiang Li
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Min Wu
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Bisheng Shi
- Department of Laboratory Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Zhong Fang
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, China
| | - Liang Chen
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, China
| | - Yumei Wen
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, China
| | - Chuanwu Zhu
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Zhu
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaonan Zhang
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Australia
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, China
| |
Collapse
|
41
|
Hu W, Gao H, Cui C, Wang L, Wang Y, Li Y, Li F, Zheng Y, Xia T, Wang S. Harnessing engineered symbionts to combat concurrent malaria and arboviruses transmission. Nat Commun 2025; 16:2104. [PMID: 40025068 PMCID: PMC11873228 DOI: 10.1038/s41467-025-57343-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 02/19/2025] [Indexed: 03/04/2025] Open
Abstract
Concurrent malaria and arbovirus infections pose significant public health challenges in tropical and subtropical regions, demanding innovative control strategies. Here, we describe a strategy that employs multifunctional engineered symbiotic bacteria to suppress concurrent transmission of malaria parasites, dengue, and Zika viruses by various vector mosquitoes. The symbiotic bacterium Serratia AS1, which efficiently spreads through Anopheles and Aedes populations, is engineered to simultaneously produce anti-Plasmodium and anti-arbovirus effector proteins controlled by a selected blood-induced promoter. Laboratory and outdoor field-cage studies show that the multifunctional engineered symbiotic strains effectively inhibit Plasmodium infection in Anopheles mosquitoes and arbovirus infection in Aedes mosquitoes. Our findings provide the foundation for the use of engineered symbiotic bacteria as a powerful tool to combat the concurrent transmission of malaria and arbovirus diseases.
Collapse
Affiliation(s)
- Wenqian Hu
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Han Gao
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chunlai Cui
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- Shanghai Institute of Wildlife Epidemics, School of Life Sciences, East China Normal University, Shanghai, China
| | - Lihua Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yiguan Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yifei Li
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Fang Li
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yitong Zheng
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Tianyu Xia
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Sibao Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
42
|
Flögel S, Strater M, Fischer D, Gründemann D. A creatine efflux transporter in oligodendrocytes. FEBS J 2025; 292:1124-1140. [PMID: 39792585 PMCID: PMC11880989 DOI: 10.1111/febs.17382] [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: 07/25/2024] [Revised: 10/14/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
Creatine is essential for ATP regeneration in energy-demanding cells. Creatine deficiency results in severe neurodevelopmental impairments. In the brain, creatine is synthesized locally by oligodendrocytes to supply neighboring neurons. Neuronal uptake is mediated by SLC6A8. However, it is still unknown how creatine is released from the producing cells. Here, we investigated the function of the transporter SLC22A15, which exhibits strikingly high amino acid sequence conservation. The release of substrates from 293 cells via heterologously expressed human and rat SLC22A15 was analyzed by mass spectrometry. A number of zwitterions were identified as substrates, with similar efflux transport efficiencies. However, in absolute numbers, the efflux of creatine far outweighed all other substrates. In contrast to the permanent creatine efflux mediated by SLC16A12 and SLC16A9, SLC22A15 was, by default, completely inactive, thereby preventing continuous creatine loss from producing cells. External substrates such as guanidinoacetic acid, GABA, or MPP+ trigger creatine release through a one-to-one exchange. Human and mouse mRNA profiles indicate that SLC22A15 expression is highest in oligodendrocytes and bone marrow. Single-cell RNA sequencing data substantiate the hypothesis that SLC22A15 depends on high intracellular creatine concentrations: high SLC22A15 counts, as in oligodendrocytes and macrophages, correlate with high counts of the creatine synthesis enzymes AGAT and GAMT in both humans and mice, whereas in proximal tubular cells and hepatocytes, AGAT counts are high, but SLC22A15 is absent. Our findings establish SLC22A15 as the pivotal transporter for controlled creatine release from oligodendrocytes, filling a critical gap in understanding creatine metabolism in the brain.
Collapse
Affiliation(s)
- Svenja Flögel
- Department of Pharmacology, Faculty of Medicine and University Hospital CologneUniversity of CologneGermany
| | - Miriam Strater
- Department of Pharmacology, Faculty of Medicine and University Hospital CologneUniversity of CologneGermany
| | - Dietmar Fischer
- Department of Pharmacology, Faculty of Medicine and University Hospital CologneUniversity of CologneGermany
| | - Dirk Gründemann
- Department of Pharmacology, Faculty of Medicine and University Hospital CologneUniversity of CologneGermany
| |
Collapse
|
43
|
Xu ZY, Gao JS, He Y, Xiao XQ, Gong GZ, Zhang M. Hepatitis B virus confers innate immunity evasion through hepatitis B virus-miR-3 down-regulation of cGAS-Sting-IFN signaling. World J Hepatol 2025; 17:99292. [PMID: 40027574 PMCID: PMC11866139 DOI: 10.4254/wjh.v17.i2.99292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/05/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) evades the innate immunity and leads to persistent chronic infection, but the molecular mechanism is still not well known. AIM To investigate whether HBV-miR-3 is involved in HBV immune evasion. METHODS HBV-miR-3 agomir and antagomir were employed to verify the effectiveness of HBV-miR-3 on cGAS-Sting-IFN pathway through the experiments on relative luciferase activity, cGAS protein expression, Sting phosphorylation and interferon (IFN) production. RESULTS HBV-miR-3 down-regulates cGAS protein expression post-transcriptionally by inhibition of cGAS 3'-untranslated region (3'-UTR) activity, which results in lower Sting phosphorylation and IFN production. HBV-miR-3 antagomir rescued cGAS protein expression, Sting phosphorylation and IFN-β production. CONCLUSION HBV-miR-3 plays an important role in HBV immunity evasion by targeting cGAS 3'-UTR and interfering with cGAS-Sting-IFN pathway.
Collapse
Affiliation(s)
- Zhen-Yu Xu
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Jia-Shi Gao
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Ying He
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xin-Qiang Xiao
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Guo-Zhong Gong
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China.
| | - Min Zhang
- Institute of Hepatology and Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
44
|
Guo J, Lu J, Du P, Cheng K, Lei C, Jiang Y, Peng M, Li Y, Sun K, Xu C, Yu Y, Gao C, Kang Q, Zhang Y, Wang R, Yang Z. Fluorescence Reduction Neutralization Test: A Novel, Rapid, and Efficient Method for Characterizing the Neutralizing Activity of Antibodies Against Dengue Virus. Curr Issues Mol Biol 2025; 47:140. [PMID: 40136394 PMCID: PMC11941698 DOI: 10.3390/cimb47030140] [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: 01/22/2025] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Dengue virus (DENV) is a major public health threat in the tropical and subtropical regions of the world. Climate change resulting from global warming is further expanding DENV-endemic areas, adversely affecting public life and health. Despite this, no specific drug against DENV has been developed so far. Vaccines and neutralizing antibodies are the chief preventive and therapeutic tools for managing pathogenic infections. The present study describes the development of a novel fluorescence reduction neutralization test (FRNT) for evaluating the neutralizing activity of antibodies against DENV. This FRNT allows rapid antibody screening. In addition, we calculated the FRNT50 to indicate the neutralizing ability of the antibodies. In contrast to the conventional plaque reduction neutralization assay, the FRNT has a shorter experimental cycle, a simpler operation, and greater objectivity, which can greatly accelerate the research and development process of vaccines and antibodies against DENV.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Rong Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (J.G.); (J.L.); (P.D.); (K.C.); (C.L.); (Y.J.); (M.P.); (Y.L.); (K.S.); (C.X.); (Y.Y.); (C.G.); (Q.K.); (Y.Z.)
| | - Zhixin Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; (J.G.); (J.L.); (P.D.); (K.C.); (C.L.); (Y.J.); (M.P.); (Y.L.); (K.S.); (C.X.); (Y.Y.); (C.G.); (Q.K.); (Y.Z.)
| |
Collapse
|
45
|
Medeiros MA, Abreu BJ, Lima JPMS. Assessing Creatine-Related Gene Expression in Kidney Disease: Can Available Data Give Insights into an Old Discussion? Nutrients 2025; 17:651. [PMID: 40004980 PMCID: PMC11858045 DOI: 10.3390/nu17040651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
The impact of creatine supplementation on individuals with kidney disease or pathological conditions with an increased risk of developing kidney dysfunction remains an active discussion. However, the literature on gene expression related to cellular creatine uptake and metabolism under altered renal function is scarce. Therefore, the present study utilized comprehensive bioinformatics analysis to evaluate the expression of creatine-related genes and to establish their relationships to normal and disturbed renal conditions. We identified 44 genes modulated explicitly in response to creatine exposure from a gene enrichment analysis, including IGF1, SLC2A4, and various creatine kinase genes. The analysis revealed associations with metabolic processes such as amino acid metabolism, indicating a connection between creatine and tissue physiology. Using the Genotype-Tissue Expression Portal, we evaluated their basal tissue-specific expression patterns in kidney and pancreas tissues. Then, we selected several pieces of Gene Expression Omnibus (GEO) transcriptomic data, estimated their expression values, and established relationships to the creatine metabolism pathways and regulation, shedding light on the potential regulatory roles of creatine in cellular processes during kidney diseases. These observations also highlight the connection between creatine and tissue physiology, emphasizing the importance of understanding the balance between endogenous creatine synthesis and creatine uptake, particularly the roles of genes such as GATM, GAMT, SLC6A8, and IGF1, under several kidney dysfunction conditions. Overall, the available data in the biological databases can provide new insights and directions into creatine's effects and role in renal function.
Collapse
Affiliation(s)
- Matheus Anselmo Medeiros
- Bioinformatics Graduate Program, Digital Metropolis Institute, Federal University of Rio Grande do Norte (UFRN), Natal 59078-400, RN, Brazil;
| | - Bento João Abreu
- Department of Morphology, Biosciences Center, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil;
| | - João Paulo Matos Santos Lima
- Bioinformatics Graduate Program, Digital Metropolis Institute, Federal University of Rio Grande do Norte (UFRN), Natal 59078-400, RN, Brazil;
- Bioinformatics Multidisciplinary Environment (BioME), Digital Metropolis Institute (IMD), Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| |
Collapse
|
46
|
Li T, Ding L, Wang Q, Ma J, Wang S. Enhancing cardiac repair post-myocardial infarction: a study on GATM/Gel hydrogel therapeutics. Cell Biol Toxicol 2025; 41:44. [PMID: 39937362 PMCID: PMC11821695 DOI: 10.1007/s10565-025-09987-5] [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: 07/01/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND AND PURPOSE Significant advancements in therapeutic approaches are imperative to address the prevalent impact of myocardial infarction (MI) on morbidity and mortality rates worldwide. This study explores the therapeutic potential of GATM/Gel hydrogel, focusing on its ability to enhance cardiac repair and functionality after MI through modulation of inflammatory and repair pathways. EXPERIMENTAL APPROACH The effects of GATM/Gel hydrogel on cardiac recovery were studied in a murine MI model. HA-CHO and gelatin solutions were mixed in situ using a dual syringe with a static mixing needle, and the resulting hydrogel was applied directly to the epicardium during MI modeling, followed by repositioning of the heart and closure of the thorax. Comprehensive in vivo assessments-including echocardiography, electrocardiography, and histopathological analysis-were combined with molecular techniques such as RT-qPCR, Western blotting, and immunofluorescence to elucidate the underlying mechanisms. Key cellular and molecular changes were tracked, focusing on macrophage polarization, angiogenesis, and modulation of the TNF/TNFR2 signaling pathway. KEY RESULTS Employing the GATM/Gel hydrogel led to a substantial improvement in heart function, shown through enhanced ejection fraction and fractional shortening, and reduced infarction size compared to control groups. Mechanistically, the hydrogel promoted the polarization of anti-inflammatory M2 macrophages and stimulated angiogenesis. Moreover, treatment with GATM/Gel hydrogel altered the TNF/TNFR2 pathway, pivotal in mediating inflammatory responses and facilitating myocardial repair. The discoveries highlight the possibility of GATM/Gel hydrogels as an innovative remedy for MI, providing a twofold role in regulating inflammation and fostering recovery.
Collapse
Affiliation(s)
- Te Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lijuan Ding
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Qiang Wang
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Jianing Ma
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Shudong Wang
- Department of Cardiology, The First Hospital of Jilin University, No. 1 Xinmin Street Avenue, Chaoyang District, Changchun, 130021, China.
| |
Collapse
|
47
|
Eskander G, Abdelhamid SG, Wahdan SA, Radwan SM. Insights on the crosstalk among different cell death mechanisms. Cell Death Discov 2025; 11:56. [PMID: 39929794 PMCID: PMC11811070 DOI: 10.1038/s41420-025-02328-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
The phenomenon of cell death has garnered significant scientific attention in recent years, emerging as a pivotal area of research. Recently, novel modalities of cellular death and the intricate interplay between them have been unveiled, offering insights into the pathogenesis of various diseases. This comprehensive review delves into the intricate molecular mechanisms, inducers, and inhibitors of the underlying prevalent forms of cell death, including apoptosis, autophagy, ferroptosis, necroptosis, mitophagy, and pyroptosis. Moreover, it elucidates the crosstalk and interconnection among the key pathways or molecular entities associated with these pathways, thereby paving the way for the identification of novel therapeutic targets, disease management strategies, and drug repurposing.
Collapse
Affiliation(s)
- Georgette Eskander
- Postgraduate program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | - Sara A Wahdan
- Pharmacology and toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
48
|
Khan A, Zakirullah, Wahab S, Hong ST. Advances in antiviral strategies targeting mosquito-borne viruses: cellular, viral, and immune-related approaches. Virol J 2025; 22:26. [PMID: 39905499 PMCID: PMC11792744 DOI: 10.1186/s12985-025-02622-z] [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: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025] Open
Abstract
Mosquito-borne viruses (MBVs) are a major global health threat, causing significant morbidity and mortality. MBVs belong to several distinct viral families, each with unique characteristics. The primary families include Flaviviridae (e.g., Dengue, Zika, West Nile, Yellow Fever, Japanese Encephalitis), transmitted predominantly by Aedes and Culex mosquitoes; Togaviridae, which consists of the genus Alphavirus (e.g., Chikungunya, Eastern and Western Equine Encephalitis viruses), also transmitted by Aedes and Culex; Bunyaviridae (recently reorganized), containing viruses like Rift Valley Fever and Oropouche virus, transmitted by mosquitoes and sometimes sandflies; and Reoviridae, which includes the genus Orbivirus (e.g., West Nile and Bluetongue viruses), primarily affecting animals and transmitted by mosquitoes and sandflies. Despite extensive research, effective antiviral treatments for MBVs remain scarce, and current therapies mainly provide symptomatic relief and supportive care. This review examines the viral components and cellular and immune factors involved in the life cycle of MBVs. It also highlights recent advances in antiviral strategies targeting host factors such as lipid metabolism, ion channels, and proteasomes, as well as viral targets like NS2B-NS3 proteases and nonstructural proteins. Additionally, it explores immunomodulatory therapies to enhance antiviral responses and emphasizes the potential of drug repurposing, bioinformatics, artificial intelligence, and deep learning in identifying novel antiviral candidates. Continued research is crucial in mitigating MBVs' impact and preventing future outbreaks.
Collapse
Affiliation(s)
- Ayyaz Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shahid Wahab
- Department of Agriculture, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea.
| |
Collapse
|
49
|
Wang YN, Liu S. The role of ALDHs in lipid peroxidation-related diseases. Int J Biol Macromol 2025; 288:138760. [PMID: 39674477 DOI: 10.1016/j.ijbiomac.2024.138760] [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: 04/15/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Lipid peroxidation presents the oxidative degradation of polyunsaturated fatty acids lincited by reactive species. Excessive accumulation of lipid peroxidation byproducts, including 4-hydroxy-2-nonenal (4-HNE) and malondialdehyde (MDA), causes protein dysfunction and various illnesses. Aldehyde dehydrogenases (ALDHs) catalyze the metabolism of both endogenous and exogenous aldehydes. These enzymes participate in detoxification and intermediary metabolism. Contemporary research has affirmed the involvement of both enzymatic and non-enzymatic pathways of ALDHs in modulating the evolution of diseases associated with lipid peroxidation. This review provides an overview of the biological functions and clinical implications concerning the enzymatic and non-enzymatic pathways of ALDHs in diseases related to lipid peroxidation, such as, non-alcoholic fatty liver disease (NAFLD), atherosclerosis, and type 2 diabetes (T2DM). Furthermore, the activators or inhibitors of ALDHs represent a promising therapeutic strategy for lipid peroxidation-related diseases.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of Implantology & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China; Suzhou Research Institute, Shandong University, Suzhou, Jiangsu 215123, China
| | - Shiyue Liu
- Department of Implantology & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China.
| |
Collapse
|
50
|
Wu D, Kao JH, Piratvisuth T, Wang X, Kennedy PT, Otsuka M, Ahn SH, Tanaka Y, Wang G, Yuan Z, Li W, Lim YS, Niu J, Lu F, Zhang W, Gao Z, Kaewdech A, Han M, Yan W, Ren H, Hu P, Shu S, Kwo PY, Wang FS, Yuen MF, Ning Q. Update on the treatment navigation for functional cure of chronic hepatitis B: Expert consensus 2.0. Clin Mol Hepatol 2025; 31:S134-S164. [PMID: 39838828 PMCID: PMC11925436 DOI: 10.3350/cmh.2024.0780] [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: 09/09/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/23/2025] Open
Abstract
As new evidence emerges, treatment strategies toward the functional cure of chronic hepatitis B are evolving. In 2019, a panel of national hepatologists published a Consensus Statement on the functional cure of chronic hepatitis B. Currently, an international group of hepatologists has been assembled to evaluate research since the publication of the original consensus, and to collaboratively develop the updated statements. The 2.0 Consensus was aimed to update the original consensus with the latest available studies, and provide a comprehensive overview of the current relevant scientific literatures regarding functional cure of hepatitis B, with a particular focus on issues that are not yet fully clarified. These cover the definition of functional cure of hepatitis B, its mechanisms and barriers, the effective strategies and treatment roadmap to achieve this endpoint, in particular new surrogate biomarkers used to measure efficacy or to predict response, and the appropriate approach to pursuing a functional cure in special populations, the development of emerging antivirals and immunomodulators with potential for curing hepatitis B. The statements are primarily intended to offer international guidance for clinicians in their practice to enhance the functional cure rate of chronic hepatitis B.
Collapse
Affiliation(s)
- Di Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Horng Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Teerha Piratvisuth
- NKC Institute of Gastroenterology and Hepatology, Songklanagarind Hospital, Prince of Songkla University, Hat Yai, Thailand
| | - Xiaojing Wang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Patrick T.F. Kennedy
- Barts Liver Centre, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Academic Fields of Medicine, Dentistry, and Pharmaceutical Science, Okayama University, Okayama, Japan
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Guiqiang Wang
- Department of Infectious Disease, Center for Liver Disease, Peking University First Hospital, Beijing, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wenhui Li
- National Institute of Biological Sciences, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Young-Suk Lim
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Junqi Niu
- Department of Hepatology, First Hospital of Jilin University, Jilin University, Jilin, China
| | - Fengmin Lu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiliang Gao
- Department of Infectious Diseases, Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Apichat Kaewdech
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Meifang Han
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Ren
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Sainan Shu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Paul Yien Kwo
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Fu-sheng Wang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Man-Fung Yuen
- Department of Medicine, School of Clinical Medicine & State Key Laboratory of Liver Research, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|