1
|
Thibaut MM, Roumain M, Piron E, Gillard J, Loriot A, Neyrinck AM, Rodriguez J, Massart I, Thissen JP, Huot JR, Pin F, Bonetto A, Delzenne NM, Muccioli GG, Bindels LB. The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia. Gut Microbes 2025; 17:2449586. [PMID: 39780051 PMCID: PMC11730681 DOI: 10.1080/19490976.2025.2449586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as Xylanibacter rodentium, as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored in vitro and in vivo. In vitro, TDCA prevented myotube atrophy, whereas in vivo hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.
Collapse
Affiliation(s)
- Morgane M. Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Edwige Piron
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Justine Gillard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Axelle Loriot
- Computational Biology and Bioinformatics Unit (CBIO), de Duve Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle Massart
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Welbio Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
Tang Y, Pan W, Ding W, Pan X, Zhu J, Chen H, Zhu X, Chen J, Cheng Z, Zhang Y, Zhang B. Prostaglandin E2 alleviates inflammatory response and lung injury through EP4/cAMP/IKK/NF-κB pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167801. [PMID: 40090625 DOI: 10.1016/j.bbadis.2025.167801] [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: 09/15/2024] [Revised: 02/10/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
PURPOSE Prostaglandin E2 (PGE2), a pivotal lipid metabolite, plays a dual role in inflammation, manifesting both pro-inflammatory and anti-inflammatory effects, which are significantly influenced by the cellular microenvironment and receptor subtype. Although recent studies have highlighted the anti-inflammatory potential of PGE2, its role in toll-like receptor (TLR)-associated inflammation and the underlying mechanisms have not fully elucidated. Consequently, the primary aim of this study was to assess the anti-inflammatory efficacy of PGE2 in TLR-related inflammation and to elucidate the associated mechanisms. METHODS In vitro, the anti-inflammatory effect of PGE2 on TLR-related inflammation were investigated by measuring pro-inflammatory cytokine protein and gene levels using ELISA and RT-qPCR, respectively. Western blot analysis was used to explore the corresponding anti-inflammatory signaling pathways. In vivo, the anti-inflammatory effects of PGE2 were further validated using ALI and sepsis models, employing the PGE2 analog 16,16-dimethyl prostaglandin E2 (dmPGE2). RESULTS The findings revealed that PGE2 inhibited the LPS-induced inflammatory response and activation of the IKK/NF-κB signaling pathway via the EP4 receptor-mediated downstream cAMP/PKA pathway. Additionally, PGE2 analog, dmPGE2, effectively mitigated pathological injury and the inflammatory response in lung tissue of mice subjected to LPS-induced ALI and sepsis. Notably, dmPGE2 suppressed LPS-induced activation of the IKK/NF-κB signaling pathway in lung tissue. CONCLUSION This study demonstrated that PGE2 can inhibit the IKK/NF-κB signaling pathway through the EP4/cAMP/PKA pathway, thereby alleviating the LPS-induced inflammatory response and providing a protective effect against LPS-induced ALI and sepsis. Consequently, PGE2 holds promise as a candidate for drug development aimed at preventing ALI and sepsis.
Collapse
Affiliation(s)
- Yelin Tang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Guangdong Food and Drug Vocational College, Guangzhou, Guangdong 510520, China
| | - Weiting Pan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Ding
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xingye Pan
- The First Affiliated Medical College of Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Junyi Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Huiwen Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaona Zhu
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
| | - Jingyi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zijun Cheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yali Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Bing Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China.
| |
Collapse
|
3
|
Romero-Gavilán F, Cerqueira A, García-Arnáez I, Scalschi L, Vicedo B, Azkargorta M, Elortza F, Izquierdo R, Gurruchaga M, Goñi I, Suay J. Proteomic evaluation of borosilicate hybrid sol-gel coatings with osteogenic, immunomodulatory and antibacterial properties. Colloids Surf B Biointerfaces 2025; 250:114561. [PMID: 39956001 DOI: 10.1016/j.colsurfb.2025.114561] [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: 12/04/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Silica hybrid sol-gel coatings represent an interesting approach to bioactivate dental implants. Boron is known for its osteogenic, angiogenic and antibacterial functions in biomedical applications. This study describes the synthesis of a novel borosilicate hybrid sol-gel coating using a mixture of methyltrimethoxysilane, tetraethyl orthosilicate and trimethyl borate (TMB). Coatings with different amounts of boron were obtained, and their physiochemical properties were examined; in vitro tests with human osteoblasts and macrophages (THP-1) were carried out. The effects of these materials on bacteria viability were evaluated using Escherichia coli and Staphylococcus aureus. The human serum proteins adsorbed onto the coatings were analysed employing proteomic techniques. To synthesise the new materials, the appropriate sol-gel reactions were developed; boron was integrated into the silica network, and well-adhering coatings were obtained. These borosilicate coatings were non-cytotoxic, displayed osteogenic potential, and upregulated adsorption of proteins related to bone regeneration (IGF2, ALS and APOE). Boron upregulated the expression of TNF-α, INFg and TGF-β and increased the TNF-α and TGF-β cytokine production in THP-1. Moreover, the addition of boron caused downregulation of NOX2 expression. Proteomic analysis revealed that boron-doping reduced the adsorption of immunoglobulins and complement system proteins. It also caused an increase in the levels of apolipoproteins, antioxidant proteins and serum amyloid A proteins, which was in agreement with in vitro results. The coatings with 10 and 20 % TMB displayed antibacterial effect against S. aureus. The results of this study will enhance our comprehension of interactions between boron-containing biomaterials and biological systems.
Collapse
Affiliation(s)
- Francisco Romero-Gavilán
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, Castellón de la Plana 12071, Spain.
| | - Andreia Cerqueira
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, Castellón de la Plana 12071, Spain
| | - Iñaki García-Arnáez
- Departament of Polymers and Advanced Materials: Physics, Chemistry and Technology, Universidad del País Vasco, P. M. de Lardizábal, 3, San Sebastián 20018, Spain
| | - Loredana Scalschi
- Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, Av. Vicent Sos Baynat s/n, Castellón de la Plana 12071, Spain
| | - Begonya Vicedo
- Department of Biology, Biochemistry and Natural Sciences, Universitat Jaume I, Av. Vicent Sos Baynat s/n, Castellón de la Plana 12071, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, Derio 48160, Spain
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, Derio 48160, Spain
| | - Raúl Izquierdo
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, Castellón de la Plana 12071, Spain
| | - Mariló Gurruchaga
- Departament of Polymers and Advanced Materials: Physics, Chemistry and Technology, Universidad del País Vasco, P. M. de Lardizábal, 3, San Sebastián 20018, Spain
| | - Isabel Goñi
- Departament of Polymers and Advanced Materials: Physics, Chemistry and Technology, Universidad del País Vasco, P. M. de Lardizábal, 3, San Sebastián 20018, Spain
| | - Julio Suay
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, Castellón de la Plana 12071, Spain
| |
Collapse
|
4
|
Wei S, Song X, Mou Y, Yang T, Wang Y, Wang H, Ren C, Song X. New insights into pathogenisis and therapies of P2X7R in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:108. [PMID: 40325043 PMCID: PMC12053563 DOI: 10.1038/s41531-025-00980-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/27/2025] [Indexed: 05/07/2025] Open
Abstract
Parkinson's disease (PD), a prevalent neurodegenerative disorder, is linked to genetics and environment, but its mechanisms remain unclear. Emerging evidence connects purinergic signaling-particularly ATP-sensitive P2X7 receptor (P2X7R)-to PD. P2X7R expression is elevated in PD patients, and its antagonist BBG mitigates 6-OHDA-induced dopaminergic neuron death. This review discusses P2X7R's structure, neural functions, PD-related mechanisms, and therapeutic potential as a targert.
Collapse
Affiliation(s)
- Shizhuang Wei
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Xiaoyu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Ting Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yao Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Hanrui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Chao Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Department of Neurology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
| |
Collapse
|
5
|
Wang J, Wang Y, Huang C, Chen Y, Li X, Jiang Z. Decursin protects against DSS-induced experimental colitis in mice by inhibiting the cGAS-STING signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5119-5127. [PMID: 39520553 DOI: 10.1007/s00210-024-03589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
While studies have shown that Angelica gigas Nakai (A. gigas) can alleviate ulcerative colitis in mice, the therapeutic role of its main active ingredient, decursin, is uncertain. Therefore, we aimed to investigate the protective effect and mechanism of decursin against inflammatory bowel disease (IBD) in vivo using mice. IBD was simulated via induction with 3% dextran sodium sulfate (DSS), with or without daily treatment with decursin (10 mg/kg or 20 mg/kg) or 5-amino salicylic acid (5-ASA; 100 mg/kg) for 14 days. Mice were weighed and monitored daily for disease activity index (DAI) scoring. Colon tissues were collected for histopathological staining analysis, and serum was collected for ELISA measurement of proinflammatory cytokines. Western blotting was employed to analyze colonic expression levels of the tight junction-related proteins ZO-1, Occludin, and Claudin 1, as well as cGAS-STING signaling pathway-associated proteins. The expression levels of major proteins were verified using immunohistochemistry and immunofluorescence. Compared with the control group, DSS-induced mice showed decreased body weight, increased DAI scores, shortening of the colon, disrupted colon tissue structure, increased serum levels of proinflammatory cytokines, increased expression of factors involved in activating the cGAS-STING signaling pathway, and reduced expression of ZO-1, Occludin, and Claudin 1. Under decursin treatment, the pathological state of IBD was less severe, proinflammatory factors were downregulated, and activation of the cGAS-STING signaling pathway was inhibited. Our findings indicate that decursin helps restore the intestinal mucosal barrier and prevents activation of the cGAS-STING signaling cascade, alleviating experimental IBD in mice.
Collapse
Affiliation(s)
- Jiamin Wang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Yudi Wang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Caisheng Huang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Yonghu Chen
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, China
| | - Zhe Jiang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, China.
| |
Collapse
|
6
|
Ghazizadeh Y, Salehi Shadkami H, Madani F, Niknam S, Adabi M. Advances in cancer nanovaccines: a focus on colorectal cancer. Nanomedicine (Lond) 2025; 20:1029-1041. [PMID: 40186876 PMCID: PMC12051617 DOI: 10.1080/17435889.2025.2486930] [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: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Nanotechnology has revolutionized cancer treatment by providing innovative solutions through nanocancer therapies, nanovaccines, and nanoparticles. This review focuses on the application of these technologies in colorectal cancer (CRC), highlighting their progression from preclinical studies to clinical trials. Nanoparticles, including liposomes, silica, gold, and lipid nanoparticles, possess unique properties that enhance drug delivery, improve therapeutic efficacy, and minimize systemic toxicity. Additionally, nanovaccines are being developed to elicit robust immune responses against CRC cells. This paper offers a comprehensive overview of the current state of nanotechnology-based treatments for CRC, emphasizing key preclinical studies and clinical trials that demonstrate their potential. Furthermore, the review discusses the challenges faced in this field. It outlines future directions for research, underscoring the need for ongoing efforts to translate these promising technologies into practical clinical applications.
Collapse
Affiliation(s)
- Yalda Ghazizadeh
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Shadkami
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Niknam
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Salehi M, Mousa Farkhani E, Moghri J, Ghasemian A, Tabatabaee SS, Hooshmand E. Global dengue fever management in health systems: identifying strategies, challenges and solutions - a scoping review protocol. BMJ Open 2025; 15:e097085. [PMID: 40280619 PMCID: PMC12035428 DOI: 10.1136/bmjopen-2024-097085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Dengue fever, the fastest-spreading mosquito-borne viral disease, poses a significant global public health challenge. Over the past two decades, its rapid spread has been driven by urbanisation, climate change and international travel, particularly affecting tropical and subtropical regions. Despite its considerable economic burden, effective antiviral treatments and vaccines remain unavailable. This study aims to bridge gaps in dengue fever management by systematically identifying and analysing strategies, challenges and solutions adopted within health systems worldwide. METHODS AND ANALYSIS This scoping review will adopt the methodological framework of Arksey and O'Malley. A comprehensive search will be conducted across databases including PubMed, Scopus, Web of Science, Embase and Cochrane Library, along with grey literature sources and manual reference list searches, covering the period from 2003 to 2024, limited to English-language publications. Search strategies will be developed using controlled vocabulary and key terms associated with various components of dengue fever management. Two independent reviewers will screen titles and abstracts based on predefined inclusion and exclusion criteria, followed by full text screening to determine final eligibility. A descriptive numerical analysis will summarise the characteristics of included studies, while a thematic analysis will provide an overview of the literature, encompassing strategies, challenges and solutions. ETHICS AND DISSEMINATION This study, approved by the Medical Ethics Committee of Mashhad University of Medical Sciences (IR.MUMS.REC.1403.142), adheres to ethical guidelines for handling publicly available data. All findings will be transparently reported and disseminated through peer-reviewed journals, relevant conferences and stakeholder engagement.
Collapse
Affiliation(s)
- Mehrdad Salehi
- Student Research Committe, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Mousa Farkhani
- Department of Epidemiology, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Moghri
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Management Sciences and Health Economics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Ghasemian
- Student Research Committe, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Saeed Tabatabaee
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Management Sciences and Health Economics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elaheh Hooshmand
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Management Sciences and Health Economics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Koren M, Zlajpah M, Poljak M, Komlos KF, Flezar MS. Expression of the stem cell markers NANOG and SOX2 in the cervical squamous carcinogenesis. Radiol Oncol 2025:raon-2025-0026. [PMID: 40272007 DOI: 10.2478/raon-2025-0026] [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: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The aim of the present study was to assess a diagnostic potential of stem cell markers NANOG and SOX2 for classifying cervical squamous intraepithelial lesions (SILs)/cervical intraepithelial neoplasia (CIN). PATIENTS AND METHODS NANOG and SOX2 expression was evaluated immunohistochemically on 40 patients: in 10 cases each of low-grade SIL (LSIL), high-grade SIL/CIN, grade 2 (HSIL/CIN 2), HSIL/CIN, grade 3 (HSIL/CIN 3), cervical squamous cell carcinoma (CSCC) and their adjacent non-dysplastic squamous epithelium. In addition, human papillomavirus (HPV) genotyping and immunohistochemical staining with p16 and Ki-67 were done. NANOG and SOX2 expression was compared between squamous lesions and controls and between squamous lesions by multiplying staining intensity (SI) by the percentage of positive cells (P) and by multiplying SI by the thickness of staining in epithelium (T) to calculate SI x P and SI x T score. RESULTS NANOG and SOX2 expression gradually increased from non-dysplastic squamous epithelium via LSIL and HSIL to CSCC. Expression of NANOG and SOX2 was higher in LSIL compared to controls (P < 0.05 for NANOG Si x P and Si x T scores and SOX2 SI x T score) and lower compared to HSIL (P < 0.05 for all SI x P and SI x T scores). HSIL/CIN 3 showed higher SOX2 expression than HSIL/CIN 2 (P < 0.05 for SI x P and SI x T scores). CONCLUSIONS Contrary to p16, NANOG and SOX2 could be effective for distinguishing LSIL from non-dysplastic changes. NANOG and SOX2 could be surrogate markers for differentiating LSIL from HSIL. Moreover, SOX2 could be helpful for distinguishing HSIL/CIN 2 from HSIL/CIN 3. Further studies with larger numbers of patients and molecular insights are needed.
Collapse
Affiliation(s)
- Miha Koren
- 1Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Margareta Zlajpah
- 1Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mario Poljak
- 2Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Kristina Fujs Komlos
- 2Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
9
|
Jackson RE, Miller G, Weild R, Atlavina S, Aylward F, Harris E, Hayward R, Godlee C. Touching the invisible: exploring intracellular host-pathogen interactions through multisensory art. Immunol Cell Biol 2025. [PMID: 40268311 DOI: 10.1111/imcb.70019] [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] [Indexed: 04/25/2025]
Abstract
The microscopic world of intracellular bacteria is rarely communicated to non-scientists. By participating in the Sensory Science Exhibition, held at St Catharine's College, University of Cambridge as part of the Cambridge Festival, we sought to address this problem by creating a 3D mammalian cell with model bacteria, including Salmonella enterica, Chlamydia trachomatis and Orientia tsutsugamushi. By hijacking eukaryotic host cellular machinery and avoiding detection, these bacteria orchestrate their survival and replication within host cells. This tactile display aimed to guide participants through key aspects of intracellular bacterial life cycles such as host cell entry, Salmonella type three secretion system (T3SS) protein secretion, O. tsutsugamushi trafficking along microtubules, and C. trachomatis replication within an inclusion. We summarize our experiences in this report. We hope our multisensory conceptualization of intracellular bacteria provided inclusive and easy-to-understand communication of complex science concepts to the general public with modalities also accessible to the low-vision and blind communities.
Collapse
Affiliation(s)
| | - Georgia Miller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Rachel Weild
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Frances Aylward
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Elizabeth Harris
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Richard Hayward
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Camilla Godlee
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Zelkoski AE, Lu Z, Sukumar G, Dalgard C, Said H, Alameh MG, Mitre E, Malloy AMW. Ionizable lipid nanoparticles of mRNA vaccines elicit NF-κB and IRF responses through toll-like receptor 4. NPJ Vaccines 2025; 10:73. [PMID: 40246950 PMCID: PMC12006303 DOI: 10.1038/s41541-025-01124-x] [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: 05/02/2024] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Ionizable lipid nanoparticles (LNP) that have enabled the success of messenger RNA (mRNA) vaccines have been shown to be immunostimulatory in the absence of mRNA. However, the mechanisms through which they activate innate immune cells is incompletely understood. Using a monocyte cell line, we compared the ability of three LNP formulations to activate transcription factors Nuclear Factor-kappa B (NF-κB) and Interferon Regulatory Factor (IRF). Comparison of signaling in knockout cell lines illustrated a role for Toll-like receptor (TLR) 4 in initiation of this signaling cascade and the contribution of the ionizable lipid component. Activation induced by empty LNPs was similar to that induced by LNPs containing mRNA, indicating that LNPs may provide the majority of innate stimulation for the mRNA vaccine platform. Our findings demonstrate that ionizable lipids within LNPs signal through TLR4 to activate NF-κB and IRF, identifying a mechanism for innate activation that can be optimized for adjuvant design.
Collapse
Affiliation(s)
- Amanda E Zelkoski
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Zhongyan Lu
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Hooda Said
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of Health Sciences, Bethesda, PA, USA
| | - Allison M W Malloy
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
11
|
Zhang X, Teng Z, Mo T, Zhao N, Ren H, Li X, Dai L, Zhang X, Qin T. Development of a chimeric 56-21 kDa antigen-based ELISA for serodiagnosis of Orientia tsutsugamushi infection. Diagn Microbiol Infect Dis 2025; 112:116841. [PMID: 40267834 DOI: 10.1016/j.diagmicrobio.2025.116841] [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: 01/27/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
At present, indirect fluorescent antibody testing (IFA) for the traditional diagnosis of scrub typhus (ST) requires high biosafety facilities to prepare whole bacterial antigens that can cross-react with similar diseases. IFA is also not suitable for large-scale epidemiological investigation. Therefore, a simple, fast, and safe serological diagnostic method with high sensitivity and specificity is needed. In this study, a chimeric protein was expressed that binds to 56-kDa proteins from Karp and Kawasaki serotypes and 21-kDa from Gilliam. The chimeric protein was used to establish an Enzyme-Linked Immunosorbent Assay (ELISA) to detect antibodies against Orientia tsutsugamushi in serum. ELISA had no cross-reaction with spot fever, murine typhus, and Q fever. Compared with IFA, the sensitivity and specificity of ELISA were 94.9 % and 97.2 %, respectively, and it detected the serotypes prevalent in China, including Karp, Gilliam, Kato, and Boryong in the initial application. The results showed that ELISA could be used for serological diagnosis of ST and large-scale serological investigation. Large-scale surveys in endemic areas can be conducted for further evaluation.
Collapse
Affiliation(s)
- Xianxian Zhang
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030000, PR China; Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, 030000, PR China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Zhongqiu Teng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Ting Mo
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030000, PR China; Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, 030000, PR China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Na Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Hongyu Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Xinting Li
- The First Affiliated Hospital of Xinjiang Medical University, Wulumuqi, 830000, PR China
| | - Lupeng Dai
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Xue Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Tian Qin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China.
| |
Collapse
|
12
|
Zhang M, Ma Z, Cui H, Miao Y, Yin Y, Wen Q, Liu Z, Huang X, Xing C, Liu K, Peng H, Song L. Involvement of circadian clock protein PER2 in controlling sleep deprivation induced HMGB1 up-regulation by targeting p300 in the cortex. Sci Rep 2025; 15:12253. [PMID: 40210902 PMCID: PMC11985928 DOI: 10.1038/s41598-025-96931-6] [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: 08/18/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Lack of sleep is a common problem in current society, which can induce various brain dysfunctions. Neuroinflammation is a typical reaction caused by sleep deficit and is considered as a common basis for various neurological disorders and cognitive impairments, but the related mechanisms have not been fully clarified. The circadian clock protein plays a critical role in maintaining physiological homeostasis, including sleep/wake cycles. Circadian disorders induced by sleep deficit might contribute to the development of neuroinflammation. In the current study, we observed that sleep deprivation (SD) induced elevated expression of High-mobility group box 1 (HMGB1), one of the most important mediators of neuroinflammation, in the cortical microglia and cerebrospinal fluids. Moreover, acetylation-dependent nuclear export of HMGB1 was involved in up-regulation and secretion of HMGB1 after sleep deprivation. Further studies indicated that sleep deprivation induced an increase in the expression of acetyltransferase p300 and a decrease in the expression of deacetylase SIRT1, which synergistically enhanced the acetylation level of HMGB1 in the cortical microglial cells, thereby triggered the nuclear export and secretion of HMGB1. Most importantly, circadian clock protein PER2 constitutively interacted with p300 and inhibited its expression in the microglial cells, which can be interrupted by PER2 downregulation upon sleep deprivation, leading to the increased expression of p300 and acetylation and secretion of HMGB1. The truncated PER2 mutant without p300 binding ability lost its ability to regulate p300 expression, indicating that PER2 functioned as a co-suppressor of p300 in regulating acetylation and expression of HMGB1. Taken together, data in this study reveal a new mechanism by which PER2 is involved in controlling HMGB1 dependent neuroinflammation induced by sleep deprivation. Maintaining PER2 levels or blocking HMGB1 acetylation in the cortex might be prospective for preventing sleep deprivation-induced neuroinflammation and the related adverse reactions in the brain.
Collapse
Affiliation(s)
- Min Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhuoyao Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Haoran Cui
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yumeng Miao
- Beijing Institute of Basic Medical Sciences, Beijing, China
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Yu Yin
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Wen
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhihui Liu
- Beijing Institute of Basic Medical Sciences, Beijing, China
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chen Xing
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Kun Liu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Peng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lun Song
- Beijing Institute of Basic Medical Sciences, Beijing, China.
- Anhui Medical University, Hefei, China.
- College of Life Science, Henan Normal University, Xinxiang, China.
- School of Pharmacy, Jiamusi University, Jiamusi, China.
| |
Collapse
|
13
|
Chiang ZC, Xu S, Zhao X, Liu M, Lin J, Chen Q. Generation and characterization of 7DC-DM1: a non-cleavable CD47-targeting antibody-drug conjugates with antitumor effects. Int J Biol Macromol 2025; 310:142844. [PMID: 40187444 DOI: 10.1016/j.ijbiomac.2025.142844] [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: 11/06/2024] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths following lung cancer in recent years. Therefore, lung or colorectal cancer therapy is very important for reducing mortality. In this study, we developed and characterized CD47-specific antibody-drug conjugates, namely 7DC-DM1 ADCs, to evaluate their therapeutic effects on lung and colorectal cancer. Both 7DC2-DM1 and 7DC4-DM1 demonstrated good binding affinities of 0.56 nM and 0.49 nM, respectively, and exhibited significant cytotoxicity, though they displayed different penetration effects. These findings suggest that the binding complexes of 7DC2-DM1 and 7DC4-DM1 with CD47 receptors adopt different conformations, leading to variations in their cellular internalized efficiencies. Molecular docking simulations revealed that 7DC2 and 7DC4 bind to CD47 molecules in distinct orientations and epitopes, differing between conserved and non-conserved regions. Furthermore, treatments with 7DC2-DM1 and 7DC4-DM1 displayed notable differences in antitumor effects in murine syngeneic tumor models derived from the MC38 cell line in C57BL/6 mice. In the tumor model treated with 7DC4-DM1, immunofluorescence staining analysis revealed a large area of necrosis in the tumor stroma, accompanied by a significant infiltration of CD11b-expressing immune cells. In summary, these results indicate that 7DC4-DM1 holds promise as a therapeutic agent for colorectal cancer treatment.
Collapse
Affiliation(s)
- Zu-Chian Chiang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China; College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, China.
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Xiangqian Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Min Liu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China.
| | - Jizhen Lin
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China.
| |
Collapse
|
14
|
Zhu Y, Cao S. Unraveling the Complexities of Myeloid-Derived Suppressor Cells in Inflammatory Bowel Disease. Int J Mol Sci 2025; 26:3291. [PMID: 40244120 PMCID: PMC11989781 DOI: 10.3390/ijms26073291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) regulate immune responses in many pathological conditions, one of which is inflammatory bowel disease (IBD), an incurable chronic disorder of the digestive tract and beyond. The pathophysiology of IBD remains unclear, likely involving aberrant innate and adaptive immunity. Studies have reported altered population of MDSCs in patients with IBD. However, their distribution varies among patients and different preclinical models of IBD. The expansion and activation of MDSCs are likely driven by various stimuli during intestinal inflammation, but the in-depth mechanisms remain poorly understood. The role of MDSCs in the pathogenesis of IBD appears to be paradoxical. In addition to intestinal inflammation, suppressive MDSCs may promote colitis-to-colon cancer transition. In this Review, we summarize recent progresses on the features, activation, and roles of MDSCs in the development of IBD and IBD-associated colon cancer.
Collapse
Affiliation(s)
| | - Siyan Cao
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
15
|
Gomez-Pinilla F, Myers SK. Traumatic brain injury from a peripheral axis perspective: Uncovering the roles of liver and adipose tissue in temperature regulation. Prog Neurobiol 2025; 247:102733. [PMID: 40032155 DOI: 10.1016/j.pneurobio.2025.102733] [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: 11/06/2024] [Revised: 01/21/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Most current treatments for TBI and other neurological disorders focus on the brain, often overlooking the significant contributions of peripheral organs to disease progression. Emerging evidence suggests that organs such as the liver and adipose tissue play crucial roles in TBI pathogenesis. The liver synthesizes lipids and proteins vital for brain function, while adipose tissue provides hormones and metabolites that influence brain activity. New research indicates that the liver and adipose tissue work in concert with the hypothalamus to regulate essential processes, such as body temperature, which become disrupted in TBI. Additionally, the brain-peripheral axis-a complex network of visceral nerve pathways, hormones, and metabolites-plays a bidirectional role in regulating brain plasticity and function. Understanding how TBI leads to dysregulation of the liver, adipose tissue, and other organs could unlock new therapeutic opportunities for treating TBI and related neurological disorders. The intricate autonomic network involving hypothalamic and enteric neurons, along with visceral nerve pathways and hormones, presents both pathological targets and therapeutic potential. We examine scientific evidence suggesting that correcting disturbances in systemic physiology could enhance the brain's capacity for healing. However, the interdependence of this autonomic network implies that treating dysfunction in one area may affect others. Therefore, we also explore the mechanisms by which diet and exercise can comprehensively impact the brain-peripheral axis, supporting the healing process. CHEMICAL COMPOUNDS: D-Fructose (PubChem CID 2723872); docosahexaenoic acid (PubChem CID 45934466); eicosapentaenoic acid (PubChem 5282847).
Collapse
Affiliation(s)
- F Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Sydney K Myers
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Gomatou G, Charpidou A, Li P, Syrigos N, Gkiozos I. Mechanisms of primary resistance to immune checkpoint inhibitors in NSCLC. Clin Transl Oncol 2025; 27:1426-1437. [PMID: 39307892 DOI: 10.1007/s12094-024-03731-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/10/2024] [Indexed: 04/16/2025]
Abstract
Immune checkpoint inhibitors (ICIs) redefined the therapeutics of non-small cell lung cancer (NSCLC), leading to significant survival benefits and unprecedented durable responses. However, the majority of the patients develop resistance to ICIs, either primary or acquired. Establishing a definition of primary resistance to ICIs in different clinical scenarios is challenging and remains a work in progress due to the changing landscape of ICI-based regimens, mainly in the setting of early-stage NSCLC. The mechanisms of primary resistance to ICIs in patients with NSCLC include a plethora of pathways involving a cross-talk of the tumor cells, the tumor microenvironment and the host, leading to the development of an immunosuppressive phenotype. The optimal management of patients with NSCLC following primary resistance to ICIs represents a significant challenge in current thoracic oncology. Research in this field includes exploring other immunotherapeutic approaches, such as cancer vaccines, and investigating novel antibody-drug conjugates in patients with NSCLC.
Collapse
Affiliation(s)
- Georgia Gomatou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andriani Charpidou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Nikolaos Syrigos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Gkiozos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Yu M, Liu G, Chen W, Qiu Y, You N, Chen S, Wei Z, Ji L, Han M, Qin Z, Sun T, Wang D. Choline metabolism in ischemic stroke: An underappreciated "two-edged sword". Pharmacol Res 2025; 214:107685. [PMID: 40054542 DOI: 10.1016/j.phrs.2025.107685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/23/2025]
Abstract
Ischemic stroke (IS) is an important cause of death and disability worldwide, but the molecular mechanisms involved are not fully understood. In this context, choline metabolism plays an increasingly important role in IS due to its multifaceted mechanisms involving neuroprotection, neuroregeneration, inflammatory response, immune regulation, and long-term health effects. With the deepening of the research on choline and its metabolites, such as trimethylamine-N-oxide (TMAO), scientists have gradually realized its key role in the occurrence, development and potential treatment of IS. This review summarizes the importance of choline in neuroprotection and long-term disease management, highlighting the complexity of choline metabolism affecting cerebrovascular health through gut microbes. Although choline and its metabolites exhibit a protective effect, excessive intake and increases in some metabolites may confer risk, suggesting the need to carefully balance dietary choline intake. The purpose of this review is to integrate the existing research results and provide a theoretical basis for further exploring the mechanism, prognosis evaluation and clinical intervention of choline metabolism in ischemic IS, hoping to provide a new perspective and enlightenment for the formulation of effective stroke prevention and treatment strategies, and promote a comprehensive understanding of heart and brain health and optimize intervention methods.
Collapse
Affiliation(s)
- Mengchen Yu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Guohao Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Wenbo Chen
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Yanmei Qiu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Nanlin You
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Sui Chen
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zhaosheng Wei
- Department of Neurosurgery, Qilu Hospital (Qingdao), Shandong University, Qingdao 266035, China
| | - Longxin Ji
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Mengtao Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Zhen Qin
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Tao Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China; Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong 253000, China.
| |
Collapse
|
18
|
Chen J, He K, Li X, Wang M, Yang Z, Wang Z, Wang K, Jiang W, Zhao L, Cui M. Overexpression of FOS enhances the malignant potential of eutopic endometrial stromal cells in patients with endometriosis‑associated ovarian cancer. Oncol Rep 2025; 53:45. [PMID: 39981914 PMCID: PMC11851058 DOI: 10.3892/or.2025.8878] [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: 10/15/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2025] Open
Abstract
Endometrial cysts of the ovary (EMC) may develop into endometriosis (EM)‑associated ovarian cancer over time (EAOC), but the pathogenesis of this disease has not been determined. In the present study, RNA sequencing was used to identify a feasible biomarker, and the molecular function of this biomarker in eutopic endometrial cells from EAOC and EMC patients was evaluated to explore the potential mechanism related to EAOC and orthotopic endometrial tissue. RNA sequencing was performed on 5 EAOC and 4 EMC tissue samples, and differential expression analysis was performed. To identify biomarkers, differentially expressed genes were subjected to protein‑protein interaction network design, Gene Ontology pathway enrichment, and Gene Set Enrichment Analysis pathway enrichment. The expression of FOS in the endometrium was detected via immunohistochemical staining. Lv‑FOS was utilized to upregulate FOS in human endometrial stromal cells (hEnSCs), and Cell Counting Kit‑8, colony formation and scratch assays were performed to assess cell viability, proliferation and migration, respectively. Western blotting was used to determine protein expression. In total, 249 genes, including FOS, were differentially expressed. Pathway enrichment analysis demonstrated that the MAPK, AP‑1, ERK and other signaling pathways were involved in the EMC‑to‑EAOC conversion. FOS upregulation in hEnSCs increased cell viability, proliferation and migration. Western blot results revealed that after FOS expression was inhibited, P21 expression was upregulated, and CDK4, Cyclin D1, p‑Stat3, MMP2 and MMP9 expression was downregulated. In conclusion, mitosis and the cell cycle were found to affect the progression of EMC to EAOC. The expression of FOS, a novel biomarker, was identified to enhance the malignant potential of eutopic endometrial stromal cells in patients with EM‑associated ovarian cancer.
Collapse
Affiliation(s)
- Junyu Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Kang He
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xin Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Mengqi Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhaoyun Yang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zeyu Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Kai Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Weiqiang Jiang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Manhua Cui
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
19
|
Pan C, Yu S, Li C, Li J, Sun P, Guo Y, Li T, Wang D, Wang K, Lyu Y, Liu X, Li X, Wu J, Zhu L, Wang H. Rapid and efficient immune response induced by a designed modular cholera toxin B subunit (CTB)-based self-assembling nanoparticle. Biomaterials 2025; 315:122946. [PMID: 39515192 DOI: 10.1016/j.biomaterials.2024.122946] [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: 08/07/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Modular self-assembling nanoparticle vaccines, represent a cutting-edge approach in immunology with the potential to revolutionize vaccine design and efficacy. Although many innovative efficient modular self-assembling nanoparticles have been designed for vaccination, the immune activation characteristics underlying such strong protection remain poorly understood, limiting the further expansion of such nanocarrier. Here, we prepared a novel modular nanovaccine, which self-assembled via a pentamer cholera toxin B subunit (CTB) domain and an unnatural trimer domain, presenting S. Paratyphi A O-polysaccharide antigen, and investigated its rapid immune activation mechanism. The nanovaccine efficiently targets draining lymph nodes and antigen-presenting cells, facilitating co-localization with Golgi and endoplasmic reticulum. In addition, dendritic cells, macrophages, B cells, and neutrophils potentially participate in antigen presentation, unveiling a dynamic change of the vaccines in lymph nodes. Single-cell RNA sequencing at early stage and iN vivo/iN vitro experiments reveal its potent humoral immune response capabilities and protection effects. This nanoparticle outperforms traditional CTB carriers in eliciting robust prophylactic effects in various infection models. This work not only provides a promising and efficient candidate vaccine, but also promotes the design and application of the new type of self-assembled nanoparticle, offering a safe and promising vaccination strategy for infection diseases.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Shujuan Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Caixia Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Juntao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Dongshu Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Kangfeng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yufei Lyu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiankai Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiang Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
20
|
Yang X, Yang S, Luo Y, Chan S, Xue M, Wang Y, Xue P, Zha C, Huang N, Xie F, Yang L, Yu R, Wang H, Lan Y, Zhang L, Jia S, Fang M. Single-cell transcriptional footprint for pseudogene SsCLEC9A is associated with antigen processing and presentation in Sus scrofa. Int J Biol Macromol 2025; 302:140629. [PMID: 39904428 DOI: 10.1016/j.ijbiomac.2025.140629] [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: 10/09/2024] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
The C-type lectin domain family 9 member A (CLEC9A) is widely recognized as the most critical receptor protein for cross presentation of dead cell associated antigens in animal dendritic cells (DCs). Surprisingly, we revealed for the first time that the sole CLEC9A (SsCLEC9A) in pigs becomes a pseudogene due to three causal mutations that occurred approximately 29.8-44.7 million years ago, challenging the significance of CLEC9A in immune cross-presentation across mammals. Interestingly, we found that SsCLEC9A can transcribe a mutated transcript encoding a truncated protein. Through fluorescence-activated cell sorting and single-cell RNA sequencing, we observed that SsCLEC9A mutant transcript is mainly expressed in DCs and correlated with the expression of its homolog CLEC7A. Further data showed that DCs with SsCLEC9A mutant transcripts exhibited reduced cellular interaction ability and downregulation of antigen presentation function, displaying the characteristics of mature DCs. In addition, introducing the conserved sequence of CLEC9A gene into FLT3L-induced bone marrow hematopoietic cells significantly increased the expression of genes involved in antigen processing and presentation. This study presents a natural mutation model of pseudogenes to understand its transcriptional adation, and provides a fundamental basis for rescuing SsCLEC9A to promote immunity in pigs in the future.
Collapse
Affiliation(s)
- Xiaoyang Yang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shaojun Yang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yabiao Luo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuheng Chan
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Mingming Xue
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yubei Wang
- Sanya Institute of China Agricultural University, Sanya 572025, China
| | - Pengxiang Xue
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chengwan Zha
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Huang
- Sanya Institute of China Agricultural University, Sanya 572025, China
| | - Fuyin Xie
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lixian Yang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Runjie Yu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Hao Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yezhi Lan
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Liguo Zhang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shangang Jia
- College of Grassland Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Sanya 572025, China.
| |
Collapse
|
21
|
Hirose T, Nakazawa H, Hattori T, Ishigaki Y, Umetsu M. A Strategy to Develop Zirconia Nanoparticle-Binding Antibodies That Can Easily Cross-Link Nanoparticles by Grafting Even Insoluble Functional Peptides. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:7225-7234. [PMID: 40064552 DOI: 10.1021/acs.langmuir.4c03545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Inorganic material-binding proteins are valuable tools for conjugating different inorganic materials. The development of efficient methods for obtaining high-affinity inorganic material-binding proteins is desirable. In this study, focusing on ZrO2, which is available in the medical field as a dental material and a nanocapsule to encapsulate anticancer substances due to its high biocompatibility, we first isolated the peptides ZrO2BPa and ZrO2BPn, which bind ZrO2 nanoparticles using the phage display technique. These peptides are insoluble alone. We prepared the variable domain of the heavy chain of heavy-chain antibodies (VHHs) with low affinity for ZrO2 nanoparticles by grafting these peptides into the complementary determining region 1 (CDR1) of cAbBCII10 VHH. The affinity for VHH was further improved by optimizing CDR3 using a phage display technique with random mutagenesis. Among the VHHs, ZrO2N3 VHH showed the highest affinity, with a KD of 1.2 × 10-7 M, showing pH-dependent binding. Mixing ZrO2-binding antibodies with ZrO2 nanoparticles improved the ZrO2 nanoparticle dispersibility in phosphate buffer, which is desirable for biological use. We also generated a bispecific antibody by fusing ZrO2-binding VHH with gold-binding VHH. Unlike chemical conjugation methods, which require complicated multistep reactions, we combined ZrO2 and Au nanoparticles by simply introducing a bispecific antibody. Thus, we demonstrated an effective method for obtaining high-affinity, inorganic material-binding VHHs and the usefulness of these VHHs as interfacial molecules.
Collapse
Affiliation(s)
- Tatsuya Hirose
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Aoba 6-6-1, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Hikaru Nakazawa
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Aoba 6-6-1, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Takamitsu Hattori
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Aoba 6-6-1, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Yuri Ishigaki
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Aoba 6-6-1, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| | - Mitsuo Umetsu
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Aoba 6-6-1, Aramaki, Aoba-ku, Sendai 980-8579, Japan
- Center for Advanced Intelligence Project, RIKEN, Chuo-ku, Tokyo 103-0027, Japan
| |
Collapse
|
22
|
Chen S, Qin Z, Zhou S, Xu Y, Zhu Y. The emerging role of intestinal stem cells in ulcerative colitis. Front Med (Lausanne) 2025; 12:1569328. [PMID: 40201327 PMCID: PMC11975877 DOI: 10.3389/fmed.2025.1569328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic idiopathic inflammatory disease affecting the colon and rectum. Characterized by recurrent attacks, UC is often resistant to traditional anti-inflammatory therapies, imposing significant physiological, psychological, and economic burdens on patients. In light of these challenges, innovative targeted therapies have become a new expectation for patients with UC. A crucial pathological feature of UC is the impairment of the intestinal mucosal barrier, which underlies aberrant immune responses and inflammation. Intestinal stem cells (ISCs), which differentiate into intestinal epithelial cells, play a central role in maintaining this barrier. Growing studies have proved that regulating the regeneration and differentiation of ISC is a promising approach to treating UC. Despite this progress, there is a dearth of comprehensive articles describing the role of ISCs in UC. This review focuses on the importance of ISCs in maintaining the intestinal mucosal barrier in UC and discusses the latest findings on ISC functions, markers, and their regulatory mechanisms. Key pathways involved in ISC regulation, including the Wnt, Notch, Hedgehog (HH), Hippo/Yap, and autophagy pathways, are explored in detail. Additionally, this review examines recent advances in ISC-targeted therapies for UC, such as natural or synthetic compounds, microbial preparations, traditional Chinese medicine (TCM) extracts and compounds, and transplantation therapy. This review aims to offer novel therapeutic insights and strategies for patients who have long struggled with UC.
Collapse
Affiliation(s)
- Siqing Chen
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhang Qin
- The Fourth Hospital of Changsha (Changsha Hospital Affiliated with Hunan Normal University), Changsha, Hunan, China
| | - Sainan Zhou
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yin Xu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
23
|
Sharma V, Fernando V, Zheng X, Choi ES, Sweef O, Thomas V, Szpendyk J, Furuta S. Immunogenic shift of arginine metabolism triggers systemic metabolic and immunological reprogramming to suppress HER2 + breast cancer. Cancer Metab 2025; 13:15. [PMID: 40114277 PMCID: PMC11927160 DOI: 10.1186/s40170-025-00384-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Arginine metabolism in tumors is often shunted into the pathway producing pro-tumor and immune suppressive polyamines (PAs), while downmodulating the alternative nitric oxide (NO) synthesis pathway. Aiming to correct arginine metabolism in tumors, arginine deprivation therapy and inhibitors of PA synthesis have been developed. Despite some therapeutic advantages, these approaches have often yielded severe side effects, making it necessary to explore an alternative strategy. We previously reported that supplementing sepiapterin (SEP), the endogenous precursor of tetrahydrobiopterin (BH4, the essential NO synthase cofactor), could correct arginine metabolism in tumor cells and tumor-associated macrophages (TAMs) and induce their metabolic and phenotypic reprogramming. We saw that oral SEP treatment effectively suppressed the growth of HER2-positive mammary tumors in animals. SEP also has no reported dose-dependent toxicity in clinical trials for metabolic disorders. In the present study, we tested our hypothesis that a long-term administration of SEP to individuals susceptible to HER2-positive mammary tumor would protect them against tumor occurrence. METHODS We administered SEP, in comparison to control DMSO, to MMTV-neu mice susceptible to HER2-positive mammary tumors for 8 months starting at their pre-pubertal stage. We monitored tumor onsets to determine the rate of tumor-free survival. After 8 months of treatment, we grouped animals into DMSO treatment with or without tumors and SEP treatment with or without tumors. We analyzed blood metabolites, PBMC, and bone marrow of DMSO vs. SEP treated animals. RESULTS We found that a long-term use of SEP in animals susceptible to HER2-positive mammary tumors effectively suppressed tumor occurrence. These SEP-treated animals had undergone reprogramming of the systemic metabolism and immunity, elevating total T cell counts in the circulation and bone marrow. Given that bone marrow-resident T cells are mostly memory T cells, it is plausible that chronic SEP treatment promoted memory T cell formation, leading to a potent tumor prevention. CONCLUSIONS These findings suggest the possible roles of the SEP/BH4/NO axis in promoting memory T cell formation and its potential therapeutic utility for preventing HER2-positive breast cancer.
Collapse
Affiliation(s)
- Vandana Sharma
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA
- Department of Zoology and Physiology, University of Wyoming, 1000 E. University Ave, Biological Science Building, Room 319F, Laramie, WY, 82071, USA
| | - Veani Fernando
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus Barbara Davis Center, Mail Stop B115, 1775 Aurora Court, Aurora, CO, 80045, USA
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Eun-Seok Choi
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Osama Sweef
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Venetia Thomas
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Justin Szpendyk
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA.
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA.
| |
Collapse
|
24
|
Shang T, Jia Z, Li J, Cao H, Xu H, Cong L, Ma D, Wang X, Liu J. Unraveling the triad of hypoxia, cancer cell stemness, and drug resistance. J Hematol Oncol 2025; 18:32. [PMID: 40102937 PMCID: PMC11921735 DOI: 10.1186/s13045-025-01684-4] [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: 12/15/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
In the domain of addressing cancer resistance, challenges such as limited effectiveness and treatment resistance remain persistent. Hypoxia is a key feature of solid tumors and is strongly associated with poor prognosis in cancer patients. Another significant portion of the development of acquired drug resistance is attributed to tumor stemness. Cancer stem cells (CSCs), a small tumor cell subset with self-renewal and proliferative abilities, are crucial for tumor initiation, metastasis, and intra-tumoral heterogeneity. Studies have shown a significant association between hypoxia and CSCs in the context of tumor resistance. Recent studies reveal a strong link between hypoxia and tumor stemness, which together promote tumor survival and progression during treatment. This review elucidates the interplay between hypoxia and CSCs, as well as their correlation with resistance to therapeutic drugs. Targeting pivotal genes associated with hypoxia and stemness holds promise for the development of novel therapeutics to combat tumor resistance.
Collapse
Affiliation(s)
- Tongxuan Shang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayi Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, 100730, China
| | - Heng Cao
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Cong
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Dongxu Ma
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiang Wang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
25
|
Rosina M, Scaricamazza S, Fenili G, Nesci V, Valle C, Ferri A, Paronetto MP. Hidden players in the metabolic vulnerabilities of amyotrophic lateral sclerosis. Trends Endocrinol Metab 2025:S1043-2760(25)00044-X. [PMID: 40090808 DOI: 10.1016/j.tem.2025.02.004] [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: 10/02/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/18/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex and rapidly progressive motor neuron disorder with a fatal outcome. Despite the remarkable progress in understanding ALS pathophysiology, which has significantly contributed to clinical trial design, ALS remains a rapidly disabling and life-shortening condition. The non-motor neuron features of ALS, including nutritional status, energy expenditure, and metabolic imbalance, are increasingly gaining attention. Indeed, the bioenergetic failure and mitochondrial dysfunction of patients with ALS impact not only the high energy-demanding motor neurons but also organs and brain areas long considered irrelevant to the disease. As such, here we discuss how considering energy balance in ALS is reshaping research on this disease, opening the path to novel targetable opportunities for its treatment.
Collapse
Affiliation(s)
- Marco Rosina
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Neuroscience, Italian National Institute of Health (ISS), Rome, Italy
| | - Silvia Scaricamazza
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Gianmarco Fenili
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Movement, Human, and Health Sciences, University of Rome 'Foro Italico', Rome, Italy
| | - Valentina Nesci
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Roma 'Tor Vergata', Rome, Italy
| | - Cristiana Valle
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Alberto Ferri
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy.
| | - Maria Paola Paronetto
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Movement, Human, and Health Sciences, University of Rome 'Foro Italico', Rome, Italy.
| |
Collapse
|
26
|
Allen PE, Adcox HE, Siff TE, Gupta S, Hunt JR, Carlyon JA. Orientia tsutsugamushi alters the intranuclear balance of cullin-1 and c-MYC to inhibit apoptosis. Infect Immun 2025; 93:e0055924. [PMID: 39976440 PMCID: PMC11895443 DOI: 10.1128/iai.00559-24] [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: 12/03/2024] [Accepted: 01/23/2025] [Indexed: 02/21/2025] Open
Abstract
Cullin-1 (Cul1), a cullin-RING ubiquitin ligase component, represses c-MYC activity in the nucleus. Orientia tsutsugamushi causes the potentially fatal rickettsiosis, scrub typhus. The obligate intracellular bacterium encodes an arsenal of ankyrin repeat-containing effectors (Anks), many of which carry a eukaryotic-like F-box motif that binds Cul1. O. tsutsugamushi reduces Cul1 levels in the nucleus. This phenomenon is not due to an alteration in Cul1 neddylation but is bacterial burden- and protein synthesis-dependent. Five of the 11 Anks capable of binding Cul1 (Ank1, Ank5, Ank6, Ank9, Ank17) sequester it in the cytoplasm when each is ectopically expressed. Ank1 and Ank6 proteins with alanine substitutions in their F-boxes that render them unable to bind Cul1 cannot exclude Cul1 from the nucleus. Coincident with the reduction of Cul1 in the nuclei of Orientia-infected cells, c-MYC nuclear levels are elevated, and Cul1 target genes are differentially expressed. Several of these genes regulate apoptosis. The resistance of O. tsutsugamushi-infected cells to staurosporine-induced apoptosis is recapitulated in uninfected cells expressing Ank1 or Ank6 but not alanine-substituted versions thereof that cannot bind Cul1. Other F-box-containing Anks that cannot bind or exclude Cul1 from the nucleus also fail to confer resistance to apoptosis. Overall, O. tsutsugamushi modulates the Cul1:c-MYC intranuclear balance as an anti-apoptotic strategy that is functionally linked to a subset of its F-box-containing Anks.
Collapse
Affiliation(s)
- Paige E. Allen
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Haley E. Adcox
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Thomas E. Siff
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Sarika Gupta
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Jason R. Hunt
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
27
|
Park SY, Pylaeva E, Bhuria V, Gambardella AR, Schiavoni G, Mougiakakos D, Kim SH, Jablonska J. Harnessing myeloid cells in cancer. Mol Cancer 2025; 24:69. [PMID: 40050933 PMCID: PMC11887392 DOI: 10.1186/s12943-025-02249-2] [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: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Cancer-associated myeloid cells due to their plasticity play dual roles in both promoting and inhibiting tumor progression. Myeloid cells with immunosuppressive properties play a critical role in anti-cancer immune regulation. Cells of different origin, such as tumor associated macrophages (TAMs), tumor associated neutrophils (TANs), myeloid derived suppressor cells (also called MDSCs) and eosinophils are often expanded in cancer patients and significantly influence their survival, but also the outcome of anti-cancer therapies. For this reason, the variety of preclinical and clinical studies to modulate the activity of these cells have been conducted, however without successful outcome to date. In this review, pro-tumor activity of myeloid cells, myeloid cell-specific therapeutic targets, in vivo studies on myeloid cell re-polarization and the impact of myeloid cells on immunotherapies/genetic engineering are addressed. This paper also summarizes ongoing clinical trials and the concept of chimeric antigen receptor macrophage (CAR-M) therapies, and suggests future research perspectives, offering new opportunities in the development of novel clinical treatment strategies.
Collapse
Affiliation(s)
- Su-Yeon Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ekaterina Pylaeva
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany
| | - Vikas Bhuria
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | | | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Rome, Italy
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-Von-Guericke University, Magdeburg, Germany
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, Essen, 45147, Germany.
- German Cancer Consortium (DKTK) Partner Site Düsseldorf/Essen, Essen, Germany.
| |
Collapse
|
28
|
Du O, Yan YL, Yang HY, Yang YX, Wu AG, Guo YK, Li K, Qiao G, Du JR, Long FY. ALPK1 signaling pathway activation by HMGB1 drives microglial pyroptosis and ferroptosis and brain injury after acute ischemic stroke. Int Immunopharmacol 2025; 149:114229. [PMID: 39933362 DOI: 10.1016/j.intimp.2025.114229] [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: 01/01/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Pyroptosis and ferroptosis emerge as remarkable contributors to neuronal death and inflammation following ischemic stroke. High mobility group box 1 (HMGB1), a principal damage-associated molecular pattern (DAMP), is implicated in pyroptosis and ferroptosis post-stroke. Our previous research has demonstrated that alpha kinase 1 (ALPK1), a novel cytoplasmic pattern recognition receptor (PRR), plays an important role in mediating inflammatory damage following ischemic stroke. However, the interaction between ALPK1 and HMGB1, and their combined impact on pyroptosis and ferroptosis post-ischemic stroke remain unexplored, which is what this study aims to investigate. Initially, we observed that ALPK1 ablation attenuated ischemic brain injury of transient middle cerebral artery occlusion (tMCAO) mice. Moreover, recombinant HMGB1 (rHMGB1) stimulation induced the greatest upregulation of ALPK1 expression in microglia compared to astrocytes and neurons. Further investigation using co-immunofluorescence, co-immunoprecipitation, pull-down assay, and molecular docking revealed an interaction between HMGB1 and ALPK1. Additionally, the exacerbation of ischemic brain injury and the induction of microglial pyroptosis and ferroptosis by rHMGB1 treatment in tMCAO mice were significantly mitigated through ALPK1 deficiency by inhibiting the NLRP3/Caspase-1/GSDMD and JAK2/STAT3 signaling pathways. The inhibitory effects of ALPK1 deficiency on pyroptosis and ferroptosis induced by rHMGB1 in microglial cells were further substantiated. Finally, glycyrrhizic acid (GA), an inhibitor of HMGB1, exhibited significant neuroprotective effects in both tMCAO mice and BV2 cells subjected to oxygen-glucose deprivation/reperfusion (OGD/R) by downregulating ALPK1 expression and inhibiting microglial pyroptosis and ferroptosis. Collectively, these findings suggest that HMGB1 may interact with ALPK1 to drive microglial pyroptosis and ferroptosis via the activation of the ALPK1/NF-κB/NLRP3/GSDMD and JAK2/STAT3 signaling pathways, thereby exacerbating brain injury following acute ischemic stroke.
Collapse
Affiliation(s)
- Ou Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Ling Yan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Han-Yinan Yang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Xin Yang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yin-Kun Guo
- Department of Radiology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuan Li
- Department of Radiology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gan Qiao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| | - Fang-Yi Long
- Laboratory Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
29
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
30
|
Ma D, Wan X, Yang H, Yang L, Peng A, Yuan Q, Li Y, Xu S. Scrub Typhus Combined With Septic Shock Disseminated Intravascular Coagulation and Significant Hyperfibrinolysis: A Case Report and Review of the Literature. Case Rep Infect Dis 2025; 2025:1931423. [PMID: 40226813 PMCID: PMC11986193 DOI: 10.1155/crdi/1931423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction: Scrub typhus is an acute infectious disease caused by Orientia tsutsugamushi, whose pathophysiology is characterized by systemic small-vessel vasculitis. Its high misdiagnosis rate stems from its nonspecific clinical features. If not diagnosed and treated in time, patients may rapidly progress to multiorgan dysfunction syndrome (MODS) or even disseminated intravascular coagulation (DIC), posing a severe threat to life. Case Presentation: The patient was a 68-year-old male with "recurrent fever and dry cough for six days." He was admitted to the hospital with a diagnosis of scrub typhus. After admission, he developed severe acute respiratory distress syndrome (ARDS), MODS, septic shock, DIC with thrombocytopenia, hypofibrinogenemia, significant hyperfibrinolysis, and myocardial depression. The patient improved following treatment with doxycycline, moxifloxacin, renal replacement therapy, blood transfusion, antifibrinolysis, invasive mechanical ventilation, and other supportive therapies. The patient's coagulation profile in DIC caused by scrub typhus demonstrated significant hyperfibrinolysis, differing from that of garden-variety sepsis, and no similar cases were identified in a search of medical literature/databases. Conclusion: The fibrinolytic system in DIC caused by scrub typhus is excessively active, and antifibrinolytic therapy may benefit such patients. Further research on the distinct coagulation abnormalities in scrub typhus-associated DIC would be highly valuable compared to sepsis-associated DIC.
Collapse
Affiliation(s)
- Dewen Ma
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - Xiaohong Wan
- Intensive Care Unit, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Haihui Yang
- Department of Cardiology, People's Hospital, Pu'er City, Yunnan, China
| | - Liying Yang
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - Ankang Peng
- Department of Cardiology, People's Hospital, Pu'er City, Yunnan, China
| | - Quping Yuan
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - You Li
- Intensive Care Unit, People's Hospital, Pu'er City, Yunnan, China
| | - Shunhang Xu
- Department of Pediatrics, People's Hospital, Pu'er City, Yunnan, China
| |
Collapse
|
31
|
Lim PN, Cervantes MM, Pham LK, Doherty SR, Tufts A, Dubey D, Mai D, Aderem A, Diercks AH, Rothchild AC. Absence of c-Maf and IL-10 enables type I IFN enhancement of innate responses to LPS in alveolar macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae029. [PMID: 40073087 PMCID: PMC11952875 DOI: 10.1093/jimmun/vkae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/19/2024] [Indexed: 03/14/2025]
Abstract
Alveolar macrophages (AMs) are lung-resident myeloid cells and airway sentinels for inhaled pathogens and environmental particles. While AMs can be highly inflammatory in response to respiratory viruses, they do not mount proinflammatory responses to all airborne pathogens. For example, we previously showed that AMs fail to mount a robust proinflammatory response to Mycobacterium tuberculosis. Here, we address this discrepancy by investigating the capacity of murine AMs for direct innate immune sensing, using LPS as a model. Use of LPS-coated fluorescent beads enabled us to distinguish between directly exposed and bystander cells to measure transcriptional responses, by RNA-sequencing after cell sorting, and cytokine responses, by flow cytometry. We find that AMs have decreased proinflammatory responses to low-dose LPS compared to other macrophage types (bone marrow-derived macrophages, peritoneal macrophages), as measured by TNF, IL-6, Ifnb, and Ifit3. The reduced response to low-dose LPS correlates with minimal TLR4 and CD14 surface expression, despite sufficient internal expression of TLR4. We also find that AMs do not produce IL-10 in response to a variety of stimuli due to low expression of the transcription factor c-Maf, while exogenous c-Maf expression restores IL-10 production in AMs. Lastly, we show that lack of IL-10 enables type I IFN enhancement of AM responses to LPS. Overall, we demonstrate AMs have a cell-intrinsic hyporesponsiveness to LPS, which makes them uniquely tolerant to low-dose exposure. Regulation of AM innate responses by distinct CD14, c-Maf, and IL-10 expression patterns has important implications for both respiratory infections and environmental airborne exposures.
Collapse
Affiliation(s)
- Pamelia N Lim
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Maritza M Cervantes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Linh K Pham
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Animal Biotechnology & Biomedical Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Sydney R Doherty
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ankita Tufts
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Divya Dubey
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alan H Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alissa C Rothchild
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
32
|
Xu J, Zhang H, Nie Z, He W, Zhao Y, Huang Z, Jia L, Du Z, Zhang B, Xia S. Cancer stem-like cells stay in a plastic state ready for tumor evolution. Neoplasia 2025; 61:101134. [PMID: 39919692 PMCID: PMC11851212 DOI: 10.1016/j.neo.2025.101134] [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: 08/01/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Cell plasticity emerges as a novel cancer hallmark and is pivotal in driving tumor heterogeneity and adaptive resistance to different therapies. Cancer stem-like cells (CSCs) are considered the root of cancer. While first defined as tumor-initiating cells with the potential to develop a heterogeneous tumor, CSCs further demonstrate their roles in cancer metastasis and adaptive therapeutic resistance. Generally, CSCs come from the malignant transformation of somatic stem cells or the de-differentiation of other cancer cells. The resultant cells gain more plasticity and are ready to differentiate into different cell states, enabling them to adapt to therapies and metastatic ecosystems. Therefore, CSCs are likely the nature of tumor cells that gain cell plasticity. However, the phenotypic plasticity of CSCs has never been systematically discussed. Here, we review the distinct intrinsic signaling pathways and unique microenvironmental niches that endow CSC plasticity in solid tumors to adapt to stressful conditions, as well as emerging opportunities for CSC-targeted therapy.
Collapse
Affiliation(s)
- Jiali Xu
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Houde Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhihao Nie
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wenyou He
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yichao Zhao
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhenhui Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
| | - Lin Jia
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China.
| | - Zhiye Du
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, China.
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Siyuan Xia
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
33
|
Yan J, Guo S, He J, Huang H, Xu Y. Myeloid-derived suppressor cells in metabolic and cardiovascular disorders. Trends Endocrinol Metab 2025:S1043-2760(25)00024-4. [PMID: 40024876 DOI: 10.1016/j.tem.2025.02.001] [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: 10/15/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
Dysregulation of immune homeostasis can precipitate chronic inflammation, thus significantly contributing to the onset and progression of metabolic and cardiovascular diseases. Myeloid-derived suppressor cells (MDSCs) constitute a heterogeneous population of immature myeloid cells that are mobilized in response to biological stressors such as tissue damage and inflammation. Although MDSCs have been extensively characterized in the contexts of cancer and infectious diseases, emerging evidence highlights their pivotal roles in the pathophysiology of metabolic and cardiovascular disorders. We discuss growing evidence for the involvement of MDSCs in the progression of metabolic and cardiovascular diseases, with the aim of deepening our understanding of MDSCs in cardiometabolic physiology and identifying the necessary steps for the development of innovative MDSC-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jingwei Yan
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Thoracic Surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Shuai Guo
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China
| | - Hanpeng Huang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Yiming Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
34
|
Li Z, Deng L, Cheng M, Ye X, Yang N, Fan Z, Sun L. Emerging role of bile acids in colorectal liver metastasis: From molecular mechanism to clinical significance (Review). Int J Oncol 2025; 66:24. [PMID: 39981904 PMCID: PMC11844338 DOI: 10.3892/ijo.2025.5730] [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: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Liver metastasis is the leading cause of colorectal cancer (CRC)‑related mortality. Microbiota dysbiosis serves a role in the pathogenesis of colorectal liver metastases. Bile acids (BAs), cholesterol metabolites synthesized by intestinal bacteria, contribute to the metastatic cascade of CRC, encompassing colorectal invasion, migration, angiogenesis, anoikis resistance and the establishment of a hepatic pre‑metastatic niche. BAs impact inflammation and modulate the immune landscape within the tumor microenvironment by activating signaling pathways, which are used by tumor cells to facilitate metastasis. Given the widespread distribution of BA‑activated receptors in both tumor and immune cells, strategies aimed at restoring BA homeostasis and blocking metastasis‑associated signaling are of importance in cancer therapy. The present study summarizes the specific role of BAs in each step of colorectal liver metastasis, elucidating the association between BA and CRC progression to highlight the potential of BAs as predictive biomarkers for colorectal liver metastasis and their therapeutic potential in developing novel treatment strategies.
Collapse
Affiliation(s)
- Zhaoyu Li
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, P.R. China
| | - Lingjun Deng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Mengting Cheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Xiandong Ye
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Nanyan Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Zaiwen Fan
- Department of Oncology, Air Force Medical Center of People's Liberation Army, Air Force Medical University, Beijing 100010, P.R. China
| | - Li Sun
- Department of Oncology, Air Force Medical Center of People's Liberation Army, Air Force Medical University, Beijing 100010, P.R. China
| |
Collapse
|
35
|
Jha K, Jaishwal P, Yadav TP, Singh SP. Self-assembling of coiled-coil peptides into virus-like particles: Basic principles, properties, design, and applications with special focus on vaccine design and delivery. Biophys Chem 2025; 318:107375. [PMID: 39674128 DOI: 10.1016/j.bpc.2024.107375] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Self-assembling peptide nanoparticles (SAPN) based delivery systems, including virus-like particles (VLP), have shown great potential for becoming prominent in next-generation vaccine and drug development. The VLP can mimic properties of natural viral capsid in terms of size (20-200 nm), geometry (i.e., icosahedral structures), and the ability to generate a robust immune response (with multivalent epitopes) through activation of innate and/or adaptive immune signals. In this regard, coiled-coil (CC) domains are suitable building blocks for designing VLP because of their programmable interaction specificity, affinity, and well-established sequence-to-structure relationships. Generally, two CC domains with different oligomeric states (trimer and pentamer) are fused to form a monomeric protein through a short, flexible spacer sequence. By using combinations of symmetry axes (2-, 3- and 5- folds) that are unique to the geometry of the desired protein cage, it is possible, in principle, to assemble well-defined protein cages like VLP. In this review, we have discussed the crystallographic rules and the basic principles involved in the design of CC-based VLP. It also explored the functions of numerous noncovalent interactions in generating stable VLP structures, which play a crucial role in improving the properties of vaccine immunogenicity, drug delivery, and 3D cell culturing.
Collapse
Affiliation(s)
- Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Thakur Prasad Yadav
- Department of Physics, Faculty of Science, University of Allahabad, Prayagraj 211002, India.
| | | |
Collapse
|
36
|
Ghazizadeh M, Khorsandi K, Najafi SMA. Synergic anti-tumor effects of photodynamic therapy and resveratrol on triple-negative breast cancer cells. Photochem Photobiol Sci 2025; 24:451-465. [PMID: 40095354 DOI: 10.1007/s43630-025-00698-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION Breast cancer is a widespread type of cancer found across the world. The use of chemotherapy in breast cancer treatment may result in side effects and the emergence of drug resistance. Hence, seeking new and efficient therapies that reduce adverse reactions is imperative. Recently, combination therapy has emerged as a fresh and innovative strategy in contrast to conventional treatment methods. Photodynamic therapy (PDT) serves as a highly effective and minimally invasive technique for addressing breast cancer, providing the option to be utilized either concurrently or in conjunction with other therapeutic approaches. Resveratrol (RES) is a polyphenol found in several food sources. Research has demonstrated that RES can inhibit cell proliferation and metastasis and trigger apoptosis in tumor cells. This research aimed to assess the impact of combining RES and photodynamic therapy on MDA-MB-231 breast cancer cells. METHODS MDA-MB-231 cells were grown in culture and subsequently exposed to different methylene blue (MB) doses while subjected to laser irradiation (PDT). Following this treatment, the cells were exposed to different RES concentrations. Cell viability was assessed utilizing the MTT assay. Light and fluorescence microscopy (AO/EB staining) were employed to observe cell morphological alterations following exposure to RES and MB-PDT. Additionally, flow cytometry was utilized to investigate cell cycle progression and apoptosis induction. RESULTS The findings indicated that the co-administration of MB-PDT and RES resulted in increased cytotoxic effects on MDA-MB-231 breast cancer cells compared to the individual application of either treatment. DISCUSSION The results of this study suggest that MB-PDT can reduce the dose and time of RES treatment and, therefore, can be indicated as a new approach for treating breast cancer cells.
Collapse
Affiliation(s)
- Masta Ghazizadeh
- Department of Cell and Molecular Biology, School of Biology, College of Sciences, University of Tehran, Tehran, Iran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.
| | - SMahmoud A Najafi
- Department of Cell and Molecular Biology, School of Biology, College of Sciences, University of Tehran, Tehran, Iran
| |
Collapse
|
37
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Wu X, Hao J, Jiang K, Wu M, Zhao X, Zhang X. Neuroinflammation and pathways that contribute to tourette syndrome. Ital J Pediatr 2025; 51:63. [PMID: 40022157 PMCID: PMC11871796 DOI: 10.1186/s13052-025-01874-3] [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: 10/20/2024] [Accepted: 01/26/2025] [Indexed: 03/03/2025] Open
Abstract
Tourette syndrome (TS), a neurological and psychological disease, typically exhibit motor and phonic tics. The pathophysiology of TS remains controversial. Currently, the recognized pathogenesis of TS is the imbalance of neurotransmitters, involving abnormality of the cortex-striatum-thalamus-cortex circuit. Recently, clinical researches demonstrate that triggers such as infection and allergic reaction could lead to the onset or exacerbation of tic symptoms. Current studies have also suggested that neural-immune crosstalk caused by inflammation is also associated with TS, potentially leading to the occurrence of tics by inducing neurotransmitter abnormalities. Herein, we review inflammation-related factors contributing to the occurrence of TS as well as the mechanisms by which immune-inflammatory pathways mediate the onset of TS. This aims to clarify the pathogenesis of TS and provide a theoretical basis for the treatment of TS.
Collapse
Affiliation(s)
- Xinnan Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Hao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Keyu Jiang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhao
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
39
|
Bolideei M, Barzigar R, Gahrouei RB, Mohebbi E, Haider KH, Paul S, Paul MK, Mehran MJ. Applications of Gene Editing and Nanotechnology in Stem Cell-Based Therapies for Human Diseases. Stem Cell Rev Rep 2025:10.1007/s12015-025-10857-0. [PMID: 40014250 DOI: 10.1007/s12015-025-10857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Stem cell research is a dynamic and fast-advancing discipline with great promise for the treatment of diverse human disorders. The incorporation of gene editing technologies, including ZFNs, TALENs, and the CRISPR/Cas system, in conjunction with progress in nanotechnology, is fundamentally transforming stem cell therapy and research. These innovations not only provide a glimmer of optimism for patients and healthcare practitioners but also possess the capacity to radically reshape medical treatment paradigms. Gene editing and nanotechnology synergistically enhance stem cell-based therapies' precision, efficiency, and applicability, offering transformative potential for treating complex diseases and advancing regenerative medicine. Nevertheless, it is important to acknowledge that these technologies also give rise to ethical considerations and possible hazards, such as inadvertent genetic modifications and the development of genetically modified organisms, therefore creating a new age of designer infants. This review emphasizes the crucial significance of gene editing technologies and nanotechnology in the progress of stem cell treatments, particularly for degenerative pathologies and injuries. It emphasizes their capacity to restructure and comprehensively revolutionize medical treatment paradigms, providing fresh hope and optimism for patients and healthcare practitioners.
Collapse
Affiliation(s)
- Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rambod Barzigar
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India
| | - Razieh Bahrami Gahrouei
- Department of Pharmacy PES College, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | - Elham Mohebbi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, Springfield, IL, USA
| | - Khawaja Husnain Haider
- Sulaiman AlRajhi Medical School, Al Bukayriyah, AlQaseem, 52726, Kingdom of Saudi Arabia
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Mohammad Javad Mehran
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India.
| |
Collapse
|
40
|
Ge Y, Zhou Q, Pan F, Wang R. Utilizing Nanoparticles to Overcome Anti-PD-1/PD-L1 Immunotherapy Resistance in Non-Small Cell Lung cancer: A Potential Strategy. Int J Nanomedicine 2025; 20:2371-2394. [PMID: 40027868 PMCID: PMC11871910 DOI: 10.2147/ijn.s505539] [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: 11/21/2024] [Accepted: 01/25/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality globally, with non-small cell lung cancer (NSCLC) constituting 85% of cases. Immune checkpoint inhibitors (ICIs) represented by anti-programmed cell death protein 1 (PD-1)/ programmed cell death ligand 1 (PD-L1) have emerged as a promising frontier in cancer treatment, effectively extending the survival of patients with NSCLC. However, the efficacy of ICIs exhibits significant variability across diverse patient populations, with a substantial proportion showing poor responsiveness and acquired resistance in those initially responsive to ICIs treatments. With the advancement of nanotechnology, nanoparticles offer unique advantages in tumor immunotherapy, including high permeability and prolonged retention(EPR) effects, enhanced drug delivery and stability, and modulation of the inflammatory tumor microenvironment(TME). This review summarizes the mechanisms of resistance to ICIs in NSCLC, focusing on tumor antigens loss and defective antigen processing and presentation, failure T cell priming, impaired T cell migration and infiltration, immunosuppressive TME, and genetic mutations. Furthermore, we discuss how nanoparticles, through their intrinsic properties such as the EPR effect, active targeting effect, shielding effect, self-regulatory effect, and synergistic effect, can potentiate the efficacy of ICIs and reverse resistance. In conclusion, nanoparticles serve as a robust platform for ICIs-based NSCLC therapy, aiding in overcoming resistance challenges.
Collapse
Affiliation(s)
- Yuli Ge
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Qiong Zhou
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Fan Pan
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Rui Wang
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| |
Collapse
|
41
|
Ko SY, Park S, Choi YH. Protocatechualdehyde Induced Breast Cancer Stem Cell Death via the Akt/Sox2 Signaling Pathway. Int J Mol Sci 2025; 26:1811. [PMID: 40076435 PMCID: PMC11899452 DOI: 10.3390/ijms26051811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Breast cancer (BC) is most frequently recognized in women and characterized by histological and molecular heterogeneity. Among the various subtypes, triple-negative BC remains the most challenging disease owing to the lack of effective molecular targets and the high frequency of breast cancer stem cells (BCSCs), which account for both recurrence and resistance to conventional treatments. Despite the availability of hormonal therapies and targeted treatments, patients still face early and late relapses, necessitating new cytotoxic and selective treatment strategies. Our study focuses on investigating the effects of protocatechualdehyde (PCA), a potent bioactive compound derived from Artemisia princeps, on CSCs in BC cells. PCA inhibited BC growth and mammosphere formation as the concentration increased. This agent decreased the fraction of the CD44+/CD24- population, the aldehyde dehydrogenase 1A-expressing population, and the protein level of Sox2 in breast CSCs by downregulating Akt and pAkt. Moreover, PCA treatment reduced the tumor volume and weight in 4T1-challenged BALB/c mice. Collectively, our findings support the anti-tumor effect of Akt/Sox2-targeting PCA, suggesting a novel utilization of PCA in BC therapy.
Collapse
Affiliation(s)
- Seung-Yeon Ko
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (S.-Y.K.); (S.P.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Seonghee Park
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (S.-Y.K.); (S.P.)
| | - Youn-Hee Choi
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea; (S.-Y.K.); (S.P.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
42
|
Zou X, Du O, Yang YR, Yang YX, Zheng ZX, Li MY, Wu AG, Du JR. Alpha protein kinase 1 knockout mitigates microglial pyroptosis and cognition deficits in ADP-heptose-stimulated mice. FASEB J 2025; 39:e70371. [PMID: 39902886 DOI: 10.1096/fj.202402162rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/10/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Microglial activation and pyroptosis are central to neuroinflammation and significantly contribute to cognitive decline associated with neurodegenerative diseases. Alpha protein kinase 1 (ALPK1) is recently identified as a critical mediator of inflammatory responses in response to ADP-heptose (a pathogen-associated molecular pattern). However, its specific role in microglial pyroptosis and cognitive dysfunction remains unclear. In this study, we investigated the effects of ALPK1 on cognitive function and pyroptosis in wild-type (WT) and ALPK1 KO mice by intracerebroventricular administration of ADP-heptose to induce neuroinflammation. Cognitive performance was evaluated using behavioral tests (the Y-Maze, Morris Water Maze, and step-down passive avoidance), while Western blot, immunofluorescence, transmission electron microscopy, and enzyme-linked immunosorbent assay were used to evaluate the expression of pyroptosis markers such as NLRP3, Caspase-1, and gasdermin D (GSDMD) in vivo and in vitro. Our results reveal that the absence of ALPK1 significantly attenuated ADP-heptose-induced cognitive deficits and neuronal injury, and inhibited the NLRP3/Caspase-1/GSDMD pathway of pyroptosis and the secretion of pro-inflammatory cytokines IL-1β and IL-18. Notably, ADP-heptose-stimulated conditioned media from primary microglial cells of ALPK1 KO mice significantly enhanced neuronal cell viability, suggesting a protective role for ALPK1 deficiency in supporting neuronal health. These findings suggest the pivotal role of ALPK1 in ADP-heptose-induced microglial pyroptosis and cognitive impairment, thereby highlighting its potential as a therapeutic target in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Xiao Zou
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Ou Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yan-Rong Yang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yu-Xin Yang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zi-Xing Zheng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Meng-Yang Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Batko K, Sączek A, Banaszkiewicz M, Małyszko J, Koc-Żórawska E, Żórawski M, Niezabitowska K, Siek K, Kraśniak A, Krzanowski M, Krzanowska K. Unravelling the Role of Serum Kallistatin on Cardiorenal Outcomes in Kidney Transplant Survivors. Kidney Blood Press Res 2025; 50:221-231. [PMID: 39933507 DOI: 10.1159/000543652] [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: 08/15/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION Kallistatin, a serine protease inhibitor, has been implicated in cardiovascular and renal protection. This study investigates its association with clinical characteristics and outcomes in long-term kidney transplant recipients (KTRs). METHODS In this longitudinal observational cohort study, we enrolled 101 KTRs between September 2016 and October 2017. The median (interquartile range) time post-transplant was 52 (36-97) months, and the follow-up time was 83 (41-85) months. All patients had documented graft function of ≥24 months and no record of acute rejection or active or chronic infection at presentation. Serum kallistatin and high-sensitivity interleukin-6 were measured at baseline using commercially available enzyme-linked immunosorbent assays. A control group of 32 healthy volunteers was also recruited. RESULTS Higher serum kallistatin levels were observed in KTRs compared to healthy controls (15.9 vs. 13.8 µg/mL; p = 0.007). Concentrations were lower in diabetic versus non-diabetic KTR (14.8 vs. 16.4 µg/mL; p = 0.021). A significant interaction between diabetic status and body mass index indicated a positive association with kallistatin levels only in diabetic KTRs (p = 0.046). Linear mixed models assessing estimated glomerular filtration rate (eGFR) change over time showed improved fit after kallistatin was included in a base model with age, sex, and baseline eGFR (p = 0.024). Cox regression showed that higher kallistatin levels were associated with an increased risk of graft loss (HR: 1.120; p = 0.049), but also remained independent of time after transplantation (HR: 1.147; p = 0.030). No association was observed for all-cause mortality. The best performance was estimated for kallistatin models adjusting for time post-transplant (c-index 0.779) and diabetic status (c-index 0.707). CONCLUSION This study highlights the complex interactions between kallistatin, renal function, and cardiometabolic status in stable, long-term KTRs. Higher kallistatin levels are associated with an increased risk of graft loss in non-diabetic patients while showing a protective effect in diabetic patients. These findings support integrated management of cardio-reno-metabolic health in KTRs.
Collapse
Affiliation(s)
- Krzysztof Batko
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
- Department of Dermatology, Jagiellonian University Medical College, Cracow, Poland
| | - Anna Sączek
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| | - Małgorzata Banaszkiewicz
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Medicine, Warsaw Medical University, Warsaw, Poland
| | - Ewa Koc-Żórawska
- 2nd Department of Nephrology, Hypertension and Internal Medicine with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland
- The Academy of Applied Medical and Social Sciences, Elblag, Poland
| | - Marcin Żórawski
- The Academy of Applied Medical and Social Sciences, Elblag, Poland
- Department of Cardiology, Lipidology and Internal Medicine with Cardiac Intensive Care Unit, Medical University of Bialystok, Bialystok, Poland
| | - Karolina Niezabitowska
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| | - Katarzyna Siek
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| | - Andrzej Kraśniak
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| | - Marcin Krzanowski
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| | - Katarzyna Krzanowska
- Department of Nephrology and Transplantology, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
44
|
Hussain Z, Zhang Y, Qiu L, Gou S, Liu K. Exploring Clec9a in dendritic cell-based tumor immunotherapy for molecular insights and therapeutic potentials. NPJ Vaccines 2025; 10:27. [PMID: 39920156 PMCID: PMC11806010 DOI: 10.1038/s41541-025-01084-2] [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: 07/18/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
The pivotal role of type 1 conventional dendritic cells (cDC1s) in the field of dendritic cell (DC)-based tumor immunotherapies has been gaining increasing recognition due to their superior antigen cross-presentation abilities and essential role in modulating immune responses. This review specifically highlights the C-type lectin receptor family 9 member A (Clec9a or DNGR-1), which is exclusively expressed on cDC1s and plays a pivotal role in augmenting antigen cross-presentation and cytotoxic T lymphocyte (CTL) responses while simultaneously mitigating off-target effects. These effects include the enhancement of the cDC1s cross-presentation, reducing autoimmune responses and systemic inflammation, as well as preventing the non-specific activation of other immune cells. Consequently, these actions may contribute to reduced toxicity and enhanced treatment efficacy in immunotherapy. The exceptional ability of Clec9a to cross-present dead cell-associated antigens and enhance both humoral and CTL responses makes it an optimal receptor for DC-based strategies aimed at strengthening antitumor immunity. This review provides a comprehensive overview of the molecular characterization, expression, and signaling mechanisms of Clec9a. Furthermore, it discusses the role of Clec9a in the induction and functional activation of Clec9a+ cDC1s, with a particular focus on addressing the challenges related to off-target effects and immune tolerance in the development of tumor vaccines. Additionally, this review explores the potential of Clec9a-targeted approaches to enhance the immunogenicity of tumor vaccines and addresses the utilization of Clec9a as a delivery target for specific agonists (such as STING agonists and αGC) to enhance their therapeutic effects. This novel approach leverages Clec9a's capacity to improve the precision and efficacy of these immunomodulatory molecules in tumor treatment. In summary, this review presents compelling evidence positioning Clec9a as a promising target for DC-based tumor immunotherapy, capable of enhancing the efficacy of vaccines and immune responses while minimizing adverse effects.
Collapse
Affiliation(s)
- Zubair Hussain
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yueteng Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Qiu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Gou
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
- China‒US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China.
| |
Collapse
|
45
|
Delgado-Almenta V, Blaya-Cánovas JL, Calahorra J, López-Tejada A, Griñán-Lisón C, Granados-Principal S. Cancer Vaccines and Beyond: The Transformative Role of Nanotechnology in Immunotherapy. Pharmaceutics 2025; 17:216. [PMID: 40006583 PMCID: PMC11859086 DOI: 10.3390/pharmaceutics17020216] [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: 12/19/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality globally, responsible for approximately 10 million deaths in 2022 and an estimated 21 million new cases in 2024. Traditional cancer treatments such as surgery, radiation therapy, and chemotherapy often present limitations in efficacy and side effects. However, immunotherapeutic vaccines have emerged as a promising approach, leveraging the body's immune system to target and eliminate cancer cells. This review examines the evolving landscape of cancer vaccines, differentiating between preventive and therapeutic strategies and highlighting the significance of tumor-specific antigens, including tumor-associated antigens (TAAs) and neoantigens. Recent advancements in vaccine technology, particularly through nanotechnology, have resulted in the development of nanovaccines, which enhance antigen stability, optimize delivery to immune cells, and promote robust immune responses. Notably, clinical data indicate that patients receiving immune checkpoint inhibitors can achieve overall survival rates of approximately 34.8 months compared to just 15.7 months for traditional therapies. Despite these advancements, challenges remain, such as the immunosuppressive tumor microenvironment and tumor heterogeneity. Emerging evidence suggests that combining nanovaccines with immunomodulators may enhance therapeutic efficacy by overcoming these obstacles. Continued research and interdisciplinary collaboration will be essential to fully exploit the promise of nanovaccines, ultimately leading to more effective and accessible treatments for cancer patients. The future of cancer immunotherapy appears increasingly hopeful as these innovative strategies pave the way for enhanced patient outcomes and an improved quality of life in oncology.
Collapse
Affiliation(s)
- Violeta Delgado-Almenta
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Jesús Calahorra
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| | - Carmen Griñán-Lisón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), Centro de Investigación Biomédica (CIBM), University of Granada, 18016 Granada, Spain
| | - Sergio Granados-Principal
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (V.D.-A.); (J.L.B.-C.); (J.C.); (A.L.-T.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, 18011 Granada, Spain
| |
Collapse
|
46
|
Cohen JR, Joubert MK, Tabassum S, Capili A, Carreon J, Xiang C, Prabhu S, Merlo A, Mytych D, Dolan DG, Kouda R. Experimental validation of a parenteral permitted daily exposure value for cleaning-induced degradants from recombinant therapeutic proteins with in vitro immunogenicity assays. J Pharm Sci 2025; 114:866-877. [PMID: 39490658 DOI: 10.1016/j.xphs.2024.10.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Multiproduct manufacturing of biotherapeutic proteins generate cleaning-induced protein degradants because of extreme pH and temperature conditions during the cleaning process. Cleaning Acceptance limits are calculated based on the maximum allowable carryover (MAC) assessment of the previously manufactured active pharmaceutical ingredient (API) - or drug product - based on the permitted daily exposure (PDE) of the previously manufactured API into the dose of subsequent product. In this study, we tested a previously determined PDE value for cleaning-induced protein degradants of 650 µg/dose. A bench-scale cleaning method was used to generate cleaning induced degradants from both a half-life extension (HLE) BiTE® molecule and a mAb product. For this investigation, degradants of HLE BiTE®-A and mAb-1 were characterized alone or after spiking of 650 µg of degradants of HLE BiTE®-A or 650 µg degradants of mAb-1, into mAb-1, respectively. These samples were characterized by endotoxin testing, size exclusion chromatography (SEC), light obscuration by HIAC, and micro-fluidic imaging (MFI). These results suggest that significant degradation of the molecule occurs because of the cleaning procedure, and it is no longer in the intact form or active state. The potential immogenic impact was assessed using a cell line assay to assess immune activation, and a human Peripheral Blood Mononuclear Cell (PBMC) assay to assess T cell activation, T cell proliferation, and cytokine release after 20 h and 7 days. Findings from the various in vitro cell-based immune activation assays suggest that the presence of 650 µg of carryover of degradants either alone or spiked into the same or a cross-product do not increase immunogenicity risk in cell-based assays - suggesting that the current PDE of 650 µg/dose for cleaning-induced degradant carryover does not have a risk of immunogenicity in patients.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States.
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Syeda Tabassum
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Allyson Capili
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Julia Carreon
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Dan Mytych
- The Department of Clinical Immunology, Amgen Inc., Thousand Oaks, CA 91320, United States
| | - David G Dolan
- The Department of Environmental Health and Safety, Amgen Inc., Thousand Oaks, CA 91320, United States
| | - Ram Kouda
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States.
| |
Collapse
|
47
|
Kang H, Jung YH, Moon J, Ryu JS, Yoon CH, Kim YH, Kim MK, Kim DH. Efficacy of RCI001 as a Therapeutic Candidate in a Primary Sjögren Syndrome Mouse Model. Cornea 2025; 44:226-233. [PMID: 39288434 PMCID: PMC11676593 DOI: 10.1097/ico.0000000000003696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/24/2024] [Accepted: 08/01/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE The aim of this study was to investigate the efficacy of RCI001 (RCI) in a mouse model of primary Sjögren syndrome. METHODS Eight 12-week-old NOD.B10-H2b mice were used in this study. All experimental animals were randomly divided into phosphate-buffered saline (PBS) and RCI groups in NOD.B10-H2b mice. The eyes of mice were topically treated with PBS or RCI twice a day for a week. Ocular surface staining (OSS) and tear secretion were compared between before and after treatment. The transcript levels of inflammatory cytokines and nicotinamide adenine dinucleotide phosphate oxidase (NOX) in the conjunctiva and cornea (CC) and lacrimal gland were assayed. In addition, immunofluorescence staining of the conjunctiva was assessed. RESULTS The RCI group showed significant clinical improvement in OSS and tear secretion after 1 week of treatment compared with the baseline (both P < 0.001) and showed better improvement in OSS and tear secretion than the PBS group after 1 week of treatment (both P < 0.05). The levels of IL-1β and IL-17 in CC and IL-6 in the lacrimal gland were also significantly reduced in the RCI group compared with the PBS group (each P < 0.05). Transcript levels of NOX2 and NOX4 were also significantly reduced in CC of the RCI group compared with those of the PBS group ( P < 0.05). The RCI group also resulted in lower conjunctival expression of oxidative stress markers (4-hydroxy-2-nonenal, hexanoyl-lysine, and NOX4) than the PBS group. CONCLUSIONS Topical RCI001 demonstrated excellent therapeutic efficacy in a mouse model of primary Sjögren syndrome by inhibiting inflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Young-ho Jung
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jayoon Moon
- Department of Ophthalmology, Saevit Eye Hospital, Goyang, Republic of Korea
| | - Jin Suk Ryu
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chang Ho Yoon
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yong Ho Kim
- RudaCure Co, Ltd, Incheon, Republic of Korea
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, Republic of Korea; and
| | - Mee Kum Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Hyun Kim
- RudaCure Co, Ltd, Incheon, Republic of Korea
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
48
|
Pei X, Ma S, Hong L, Zuo Z, Xu G, Chen C, Shen Y, Liu D, Li C, Li D. Molecular insights of T-2 toxin exposure-induced neurotoxicity and the neuroprotective effect of dimethyl fumarate. Food Chem Toxicol 2025; 196:115166. [PMID: 39617286 DOI: 10.1016/j.fct.2024.115166] [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: 10/27/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
T-2 toxin, a potent environmental pollutant, has been proved to stimulate neuroinflammation, while the connection between T-2 toxin and pyroptosis remain elusive. Dimethyl fumarate (DMF), recently identified as a neuroprotectant and pyroptosis inhibitor, has potential therapeutic applications that are underexplored. Based on present study in vitro and vivo, we demonstrated that T-2 toxin induced the activation of NLRP3-Caspase-1 inflammasome in hippocampal neurons. In addition to proinflammatory mediator overexpression, gasdermin D (GSDMD)-dependently pyroptosis in the mouse hippocampal neuron cell line (HT22) treated by T-2 toxin was determined in our study. Moreover, the palliative effect of knockdown sequence of high mobility group B1 protein (HMGB1) provided more details for T-2 toxin-initiated pyroptosis. Importantly, we confirmed that DMF, as a novel inhibitor of GSDMD, could alleviate pyroptosis induced by T-2 toxin in an GSDMD targeting manner. In summary, our studies exposed the evidence that T-2 toxin could induce NLRP3 inflammasome activation and hippocampal neuronal pyroptosis. More notably, DMF was turn out to be a critical executioner for attenuating GSDMD-mediated pyroptosis. Our data found a new function of DMF and suggested a novel therapy strategy against mycotoxin-triggered neuronal inflammation, which leads to varieties of neurological diseases.
Collapse
Affiliation(s)
- Xingyao Pei
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Shuhui Ma
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Liang Hong
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Zonghui Zuo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Gang Xu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Chun Chen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Yao Shen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Dingkuo Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China
| | - Cun Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Daowen Li
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China.
| |
Collapse
|
49
|
Song H, Yoo SJ, Park WJ, Cho S, Park KS, Sung JH, Park SJ, Yoon SY, Kim K, Choi DP, Kim HM, Ju B, Youn K. List of occupational diseases among farmers in Korea: a literature review. Ann Occup Environ Med 2025; 37:e2. [PMID: 39967337 PMCID: PMC11849992 DOI: 10.35371/aoem.2025.37.e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
A comprehensive list of occupational diseases among farmers is crucial for both compensation and prevention efforts. In Korea, most farmers are self-employed, and some occupational diseases are compensated through farmer safety insurance. However, it is not harmonized with industrial accident compensation insurance and does not adequately reflect the true burden of occupational diseases among farmers. To address this gap, the authors compiled a list of occupational diseases tailored to Korean farmers by reviewing the International Labor Organization's list of occupational diseases, the Korean Industrial Accident Compensation Insurance List, the occupational disease lists of other countries, and relevant literature on farmers' work-related diseases.
Collapse
Affiliation(s)
- Hansoo Song
- Department of Occupational and Environmental Medicine, Chosun University Hospital, Gwangju, Korea
| | - Seok-Ju Yoo
- Department of Preventive Medicine, Dongguk University College of Medicine, Gyeongju, Korea
| | - Won-Ju Park
- Department of Occupational and Environmental Medicine, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Seunghyeon Cho
- Department of Occupational and Environmental Medicine, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Ki Soo Park
- Department of Preventive Medicine & Institute of Medical Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Joo Hyun Sung
- Department of Occupational and Environmental Medicine, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Sang Jin Park
- Department of Occupational and Environmental Medicine, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Seong-yong Yoon
- Department of Occupational and Environmental Medicine, Soonchunhyang University Gumi Hospital, Gumi, Korea
| | - Kyeongsoo Kim
- Agricultural Safety and Health Division, Department of Agricultural Engineering, National Institute of Agricultural Science, Rural Development Administration, Jeonju, Korea
| | - Dong-phil Choi
- Agricultural Safety and Health Division, Department of Agricultural Engineering, National Institute of Agricultural Science, Rural Development Administration, Jeonju, Korea
| | - Hye-min Kim
- Department of Occupational and Environmental Medicine, Chosun University Hospital, Gwangju, Korea
| | - Bounggyun Ju
- Department of Occupational and Environmental Medicine, Chosun University Hospital, Gwangju, Korea
| | - Kanwoo Youn
- Department of Occupational & Environmental Medicine, Wonjin Green Hospital, Seoul, Korea
| |
Collapse
|
50
|
Dow CT, Kidess Z. Proposing Bromo-epi-androsterone (BEA) for perioperative neurocognitive disorders with Interleukin-6 as a druggable target. J Clin Anesth 2025; 101:111736. [PMID: 39746239 DOI: 10.1016/j.jclinane.2024.111736] [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: 10/23/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Cognitive impairment following surgery is a significant complication, affecting multiple neurocognitive domains. The term "perioperative neurocognitive disorders" (PND) is recommended to encompass this entity. Individuals who develop PND are typically older and have increases in serum and brain pro-inflammatory cytokines notwithstanding the type of surgery undergone. Surgical trauma induces production of small biomolecules, damage-associated molecular patterns (DAMP), particularly the DAMP known as high molecular group box 1 protein (HMGB1). Mechanistically, peripheral surgical trauma promotes pro-inflammatory cytokines that stimulate central nervous system (CNS) inflammation by disrupting the blood-brain barrier (BBB) causing functional neuronal disruption that leads to PND. PND is strongly linked to elevations in serum and CNS pro-inflammatory cytokines interleukin-1 beta (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα); these cytokines cause further release of HMGB1 creating a positive feedback loop that amplifies the inflammatory response. The cytokine IL-6 is necessary and sufficient for PND. Dehydroepiandrosterone (DHEA) is a principal component of the steroid metabolome and is involved in immune homeostasis. DHEA has been shown to suppress expression of several pro-inflammatory cytokines by regulation of the NF-kB pathway. Bromo-epi-androsterone (BEA) is a potent synthetic analog of DHEA; unlike DHEA, it is non-androgenic, non-anabolic and is an effective modulator of immune dysregulation. In a randomized, placebo-controlled clinical trial, BEA effected significant and sustained decreases in IL-1β, TNFα and IL-6. This article presents BEA as a potential candidate for clinical trials targeting PND and further suggests the use of BEA in elective total hip arthroplasty as a well-documented surgical entity relevant to the management of PND.
Collapse
Affiliation(s)
- Coad Thomas Dow
- McPherson Eye Research Institute, University of Wisconsin-Madison, 9431 Wisconsin Institutes for, Medical Research (WIMR), 1111 Highland Avenue, Madison, WI 53705, United States of America.
| | - Zade Kidess
- Department of Chemistry and Biochemistry, Creighton University, 2500 California Plaza, Omaha, NE 68178, United States of America.
| |
Collapse
|