101
|
Peng Y, Lin C, Zhang B, Yan L, Zhang B, Zhao C, Qiu L. Characteristics and preliminary immune function of SRA5 in Lateolabrax maculatus. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110266. [PMID: 40064212 DOI: 10.1016/j.fsi.2025.110266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
Scavenger receptors (SRs) are crucial for pattern recognition in the innate immune system. However, the role of Scavenger Receptors class A member 5 (SRA5) in the immunological response of bony fish to pathogen invasion remains unclear. This study identified and characterized the SRA5 of Lateolabrax maculatus (LmSRA5) from its transcriptome database. LmSRA5 has a 1494 bp open reading frame, encodes 497 amino acids, has a molecular weight of 55.01 kDa, and contains a collagen domain and a conserved Scavenger Receptor Cysteine-Rich domain. LmSRA5 exhibited high sequence similarity to previously reported SRA5 genes. LmSRA5 exhibited high sequence similarity to previously reported SRA5 genes. LmSRA5 is primarily localized in the cytoplasm, with its encoded proteins distributed in both the cytoplasm and the cell membrane. LmSRA5 was expressed in all tissues. The highest expression was observed in the pituitary gland, with significant levels in the stomach, intestines, liver, and kidney. LmSRA5 expression in the head kidney, spleen, blood, and intestines initially increased, then decreased following infection with Aeromonas veronii. The binding affinity of LmSRA5 for A. veronii was enhanced by increasing concentrations of the extracellular domain recombinant LmSRA5. Knockdown and overexpression experiments in liver cells demonstrated that LmSRA5 significantly regulates the expression of IL-8 and c-Jun. LmSRA5 participates in the immune response by recognizing pathogen-associated molecular patterns (PAMPs) and contributes to immune regulation through modulation IL-8 and c-Jun. This study offers valuable insights into the role of SRA5 in pathogen resistance and immune regulation in bony fish, thereby contributing to the advancement of aquaculture under escalating disease pressures.
Collapse
Affiliation(s)
- Yangtao Peng
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China
| | - Changhong Lin
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China.
| | - Lulu Yan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China.
| | - Chao Zhao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China
| | - Lihua Qiu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China; Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Chinese Academy of Fishery Science, Beijing, PR China.
| |
Collapse
|
102
|
Dong D, Yu X, Liu H, Xu J, Guo J, Guo W, Li X, Wang F, Zhang D, Liu K, Sun Y. Study of immunosenescence in the occurrence and immunotherapy of gastrointestinal malignancies. Semin Cancer Biol 2025; 111:16-35. [PMID: 39929408 DOI: 10.1016/j.semcancer.2025.01.007] [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: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/25/2025]
Abstract
In human beings heterogenous, pervasive and lethal malignancies of different parts of the gastrointestinal (GI) tract viz., tumours of the oesophagus, stomach, small intestine, colon, and rectum, represent gastrointestinal malignancies. Primary treatment modality for gastric cancer includes chemotherapy, surgical interventions, radiotherapy, monoclonal antibodies and inhibitors of angiogenesis. However, there is a need to improve upon the existing treatment modality due to associated adverse events and the development of resistance towards treatment. Additionally, age has been found to contribute to increasing the incidence of tumours due to immunosenescence-associated immunosuppression. Immunosenescence is the natural process of ageing, wherein immune cells as well as organs begin to deteriorate resulting in a dysfunctional or malfunctioning immune system. Accretion of senescent cells in immunosenescence results in the creation of a persistent inflammatory environment or inflammaging, marked with elevated expression of pro-inflammatory and immunosuppressive cytokines and chemokines. Perturbation in the T-cell pools and persistent stimulation by the antigens facilitate premature senility of the immune cells, and senile immune cells exacerbate inflammaging conditions and the inefficiency of the immune system to identify the tumour antigen. Collectively, these conditions contribute positively towards tumour generation, growth and eventually proliferation. Thus, activating the immune cells to distinguish the tumour cells from normal cells and invade them seems to be a logical strategy for the treatment of cancer. Consequently, various approaches to immunotherapy, viz., programmed death ligand-1 (PD-1) inhibitors, Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors etc are being extensively evaluated for their efficiency in gastric cancer. In fact, PD-1 inhibitors have been sanctioned as late late-line therapy modality for gastric cancer. The present review will focus on deciphering the link between the immune system and gastric cancer, and the alterations in the immune system that incur during the development of gastrointestinal malignancies. Also, the mechanism of evasion by tumour cells and immune checkpoints involved along with different approaches of immunotherapy being evaluated in different clinical trials will be discussed.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Haoran Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiayan Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Fei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Dongyong Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Kaiwei Liu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
103
|
Jia ZX, Xiao BT, Li J, Cai XH, Qin W, Zhou M, Lu XZ. BTK inhibitors enhance NKG2D ligand expression by regulating IL-10/STAT3 pathway in activated non-GCB diffuse large B-cell lymphoma cells. Anticancer Drugs 2025; 36:374-382. [PMID: 40029697 DOI: 10.1097/cad.0000000000001696] [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] [Indexed: 03/05/2025]
Abstract
The aim of this study is to explore the role of the IL-10/STAT3 pathway in the upregulation of natural killer group 2, member D (NKG2D) ligands (MICA and ULBP2) induced by Bruton's tyrosine kinase (BTK) inhibitors in non-germinal center B-cell-like diffuse large B-cell lymphoma cells. The expression levels of NKG2D ligands and the IL-10/STAT3 pathway in SUDHL4, U2932, and OCI-LY3 cells were analyzed using western blotting. After stimulation of the B-cell receptor signaling pathway with IgM antibodies, the expression levels of NKG2D ligands, as well as IL-10 and phosphorylated STAT3 (p-STAT3) were significantly reduced. In contrast, treatment with ibrutinib produced effects opposite to those induced by IgM antibodies. Additionally, treatment of U2932 and OCI-LY3 cells with the STAT3 inhibitor (STAT3-IN-1) led to an increase in NKG2D ligand expression and a decrease in IL-10 levels. When IL-10 neutralizing antibodies were introduced, p-STAT3 levels decreased, and NKG2D ligand expression increased. Similar outcomes were observed when the BTK inhibitors ACP-196 and BGB-3111 were administered. Our findings suggest that the IL-10/STAT3 pathway plays a key role in the upregulation of NKG2D ligands induced by BTK inhibitors in U2932 and OCI-LY3 cells.
Collapse
Affiliation(s)
- Zhu-Xia Jia
- Department of Hematology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University
- The Changzhou Medical Center of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Bi-Tao Xiao
- Department of Hematology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University
| | - Jin Li
- Department of Hematology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University
| | - Xiao-Hui Cai
- Department of Hematology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University
| | - Wei Qin
- Department of Hematology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University
| | - Min Zhou
- Department of Hematology, The Third People's Hospital of Changzhou
| | - Xu-Zhang Lu
- Department of Hematology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University
- The Changzhou Medical Center of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
104
|
Ahmadizad Firouzjaei A, Aghaee-Bakhtiari SH. Integrating cuproptosis and immunosenescence: A novel therapeutic strategy in cancer treatment. Biochem Biophys Rep 2025; 42:101983. [PMID: 40224540 PMCID: PMC11986980 DOI: 10.1016/j.bbrep.2025.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/01/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Recent advancements in our understanding of cell death mechanisms have progressed beyond traditional apoptosis to encompass various forms of regulated cell death, notably cuproptosis. This copper-dependent cell death occurs when copper interacts with lipoylated enzymes in the tricarboxylic acid cycle, leading to protein aggregation and subsequent cell death. Alongside this, immunosenescence the gradual decline in immune function due to aging has emerged as a significant factor in cancer progression and response to treatment. Innovative strategies that integrate cuproptosis and immunosenescence are showing considerable promise in cancer therapy. By leveraging the altered copper metabolism in cancer cells, cuproptosis can selectively induce cell death, effectively targeting and eliminating tumors. Simultaneously, addressing immunosenescence can rejuvenate the aging immune system, enhancing its capacity to identify and destroy cancer cells. This dual approach creates a synergistic effect, optimizing therapeutic efficacy by directly attacking tumor cells while revitalizing the immune response. Such integration bolsters the defense against cancer progression and recurrence and holds great potential for advancing cancer treatment modalities and improving patient outcomes. This paper delves into the interactions between cuproptosis and immunosenescence, emphasizing their implications for developing innovative cancer therapies.
Collapse
Affiliation(s)
- Ali Ahmadizad Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
105
|
Lv Z, Xu C, Wang Z, Liu Z, Jiao J. Excessive activation?induced cytidine deaminase accumulated by proteasome inhibitors rescues abnormal class switch in activated B?cell?like diffuse large B?cell lymphoma. Exp Ther Med 2025; 29:113. [PMID: 40242597 PMCID: PMC12000863 DOI: 10.3892/etm.2025.12863] [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: 07/17/2024] [Accepted: 10/18/2024] [Indexed: 04/18/2025] Open
Abstract
Activation-induced cytidine deaminase (AID) is an enzyme that plays a crucial role in mediating somatic hypermutation and class-switch recombination (CSR). It has been found to be associated with aberrant immunoglobulin CSR in activated B-cell-like diffuse large B-cell lymphoma (ABC-DLBCL). In the present study, MG132, a potent proteasome and calpain inhibitor, induced significant cell death in ABC-DLBCL cells and inhibited the growth of ABC-DLBCL cell xenograft tumors. The results also showed that MG132 induced AID accumulation by impairing proteasome degradation of AID. Excessive endogenous AID accumulation was observed in both AID-deficient and C57/BL6 wild-type mice treated with MG132, and apparent CSR of IgM to IgG1, IgG3 and IgE. Upon stimulation of cytokines such as LPS and/or IL-4, ABC-DLBCL cells also showed a noticeable increase in CSR of IgM to IgG1, IgG3 and IgE with decreased AID protein levels. The present study demonstrates that MG132 can induce AID accumulation, which in turn restores dysfunctional CSR in ABC-DLBCL. Using MG132 as a tool, the present study elucidates the anti-lymphoma effect of proteasome inhibitors on ABC-DLBCL by rescuing the abnormal AID-induced CSR.
Collapse
Affiliation(s)
- Zhuangwei Lv
- Department of Laboratory Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Chen Xu
- Department of Infectious Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhenzhen Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zixian Liu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Junna Jiao
- Department of Laboratory Medicine, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
106
|
Qu P, Yuan J, Wu Y, Tian S, Wu Z, Chen P, Pan M, Weng H, Mai K, Zhang W. Yellow mealworm (Tenebrio molitor) meal replacing dietary fishmeal alters the intestinal microbiota, anti-oxidation and immunity of large yellow croaker (Larimichthyscrocea). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110272. [PMID: 40081435 DOI: 10.1016/j.fsi.2025.110272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
The present study investigated the impact of dietary fishmeal replacement with yellow mealworm (Tenebrio molitor) meal (TM) on the anti-oxidation, immunity and intestinal microbiota of large yellow croaker (Larimichthys crocea), with an initial body weight of 189.18 ± 0.13 g. Seven isonitrogenous and isolipidic diets were made using TM replacing the fishmeal at 0 % (TM0), 15 % (TM15), 30 % (TM30), 45 % (TM45), 60 % (TM60), 75 % (TM75) and 100 % (TM100), respectively. Each experimental diet was randomly assigned to three replicate groups of large yellow croaker (100 fish per group). After an 80-day feeding trial, it was showed that the activities of maltase, lysozyme and acid phosphatase in intestine, as well as serum albumin concentration and total anti-oxidative capacity in serum of fish fed diets with TM replacing fishmeal more than 60 % (P < 0.05). The intestinal muscularis thickness and perimeter-to-diameter ratio decreased significantly in TM75 and TM100 groups compared to the control (P < 0.05). At the transcriptional level, the mRNA expression of occludin, zonula occludens-1 (zo-1), oligopeptide transporter 1 (pept1), and oligopeptide transporter 2 (pept2) in the intestine, along with nuclear factor erythroid 2-related factor 2 (nrf2) and interleukin-10 (il-10) in both the intestine and liver, were linearly downregulated as the fishmeal replacement level increased (P < 0.05). While the relative expression levels of kelch-like ECH-associated protein 1 (keap-1), nuclear factor-κB (nf-κb) and tumor necrosis factor-α (tnf-α) in the intestine and liver were linearly upregulated with increasing dietary fishmeal replacement levels (P < 0.05). Besides, increasing dietary fishmeal replacement levels maintained intestinal microbiota alpha diversity (P > 0.05), while altering the intestinal microbial composition (P < 0.05). In conclusion, replacing 45 % of fishmeal with TM (equivalent to 25.57 % TM in diet) had no significant negative effects on the intestinal microflora, anti-oxidation and immunity of large yellow croaker.
Collapse
Affiliation(s)
- Peng Qu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Jing Yuan
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Yang Wu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Shuangjie Tian
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Zhenhua Wu
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Peng Chen
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Mingzhu Pan
- Department of Ocean Technology, College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Huasong Weng
- Ningde Fufa Fisheries Co. Ltd, Ningde, 352100, China
| | - Kangsen Mai
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Mariculture (Ministry of Education), The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), Fisheries College, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
107
|
Altun S, Özdemir S, Arslan H, Kiliçlioğlu M, Yaprak E, Bolat İ, Aydın Ş. Impact of long-term deltamethrin exposure on Alzheimer's-related neurodegeneration in rats. Exp Neurol 2025; 388:115223. [PMID: 40120660 DOI: 10.1016/j.expneurol.2025.115223] [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: 11/14/2024] [Revised: 02/27/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
In recent years, epidemiological studies have emerged indicating a potential association between chronic exposure to pesticides and the development of chronic neurodegenerative nervous system diseases such as Alzheimer's disease. In this study, we aimed to investigate the potential role of long-term nonfatal exposure to Deltamethrin in spreading this disease. To this end, a range of aspects of brain damage were discussed in rats administered deltamethrin in oral doses of 0.65 mg/kg b.w. and 1.3 mg/kg b.w. for 30 days. The activation of beta-amyloid, the primary component of plaques characteristic of Alzheimer's disease, and the NG2, a type 1 transmembrane protein, was assessed by immunohistochemistry and western blot methods in rat brain. In addition, the expression level of the APP, GFAP, NfL, TNF-alpha, CXCL9, CCL5, and IL-1 alpha genes in deltamethrin-exposed brain tissue was measured using qRT-PCR. In addition, levels of pTau181 and Abeta42 were measured with ELISA. A strong positive immunohistochemical reaction for beta-amyloid was detected in the deltamethrin-exposed brain tissues. A decrease in NG2 immunofluorescence positivity was found in the application groups compared to the control group. It was demonstrated that deltamethrin exposure significantly up-regulated the expressions of APP, GFAP, NfL, TNF-alpha, CXCL9, CCL5, and IL-1 alpha genes, also significantly higher the levels of pTau181 and Abeta42 (pg/ml) in rat brain tissues. This study provides scientific evidence that exposure to chronic doses of deltamethrin may play a positive role in the development of diseases such as Alzheimer's. Future studies should investigate similar projects and expand knowledge on the topic.
Collapse
Affiliation(s)
- Serdar Altun
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey.
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Harun Arslan
- Department of Basic Sciences, Faculty of Fisheries, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Metin Kiliçlioğlu
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Esra Yaprak
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Yakutiye, 25240 Erzurum, Turkey
| | - İsmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Şeyma Aydın
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| |
Collapse
|
108
|
Li S, Huang Y, Sun Q, Li Y, Xie H, Fu Q. Caspase-1 is critical for mice in the defense against Streptococcus equi subsp. zooepidemicus infection by promoting macrophage phagocytosis. Microb Pathog 2025; 203:107499. [PMID: 40122410 DOI: 10.1016/j.micpath.2025.107499] [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: 07/29/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an important pathogen which is responsible for a wide range of diseases in various species. Macrophages are professional phagocytes that can engulf microorganisms and trigger responses leading to microbial death. Caspase-1 is considered as a proinflammatory factor that mediates antibacterial response to protect hosts from bacteria. Here, we revealed a novel role of Caspase-1 in mice against SEZ. Through both in vitro and in vivo infection assays, we demonstrated that the maturation and secretion of the cytokine IL-1β are critically dependent on Caspase-1 activation. The Caspase-1 deficient mice displayed attenuation of bactericidal activity against SEZ, mainly by decreasing the accumulation of macrophage. In addition to the recruitment of macrophages, deficiency of Caspase-1 also impaired the phagocytosis of SEZ by macrophages. Our study demonstrated that Caspase-1 is critical for mice to defense against SEZ depending on the recruitment and phagocytosis of macrophage.
Collapse
Affiliation(s)
- Shun Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yunfei Huang
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Qinqin Sun
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yajuan Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Honglin Xie
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| | - Qiang Fu
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| |
Collapse
|
109
|
Haas G, Seiler M, Nguyen J, Troxler L, Pennarun S, Lefebvre E, Benamrouche Y, Loizeau L, Reinbolt C, Liang M, Lin X, Li W, Xia Z, Marques JT, Imler JL. Regulation of detoxifying enzymes expression and restriction of picorna-like virus infection by natural polysaccharide extracts in Drosophila cells. Virology 2025; 607:110513. [PMID: 40163969 DOI: 10.1016/j.virol.2025.110513] [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: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The world is currently witnessing a rise in viral infections, while the availability of antiviral drugs remains limited. Traditional Chinese medicine (TCM) has historically served as a valuable source of novel compounds for disease treatment. In this study, we assessed the antiviral potential of various TCM compounds using Drosophila melanogaster as a model organism. Our findings reveal that natural polysaccharide extracts, prepared from 10 commonly used medicinal herbs or fungi, exhibit antiviral activity against two picorna-like viruses. Importantly, the antiviral effect is not directly attributable to the compound itself but is instead mediated by cellular responses induced by treatment with the extract. We observed that the polysaccharide extract triggers a broad transcriptional response, which partially overlaps with NF-κB pathway activation in Drosophila. However, the antiviral activity of the extract was independent of classical innate immune pathways, such as RNA interference or NF-κB signaling. Instead, the extract appears to uniquely stimulate detoxification pathways, including upregulation of cytochrome P450 and glutathione S-transferase genes, which correlates with its antiviral effects.
Collapse
Affiliation(s)
- Gabrielle Haas
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Mélodie Seiler
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Jenny Nguyen
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Laurent Troxler
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Samuel Pennarun
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Elise Lefebvre
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | | | - Loriane Loizeau
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Cody Reinbolt
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Ming Liang
- Infinitus (China) Company Ltd., Guangzhou, China
| | | | - Wenzhi Li
- Infinitus (China) Company Ltd., Guangzhou, China
| | - Zumeng Xia
- Infinitus (China) Company Ltd., Guangzhou, China
| | - Joao T Marques
- Université de Strasbourg, INSERM U1257, CNRS UPR9022, 67084, Strasbourg, France; Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France.
| |
Collapse
|
110
|
Charnsatabut C, Suwanchaikasem P, Rattanapisit K, Iksen I, Pongrakhananon V, Bulaon CJI, Phoolcharoen W. Optimized expression of human interleukin-15 in Nicotiana benthamiana and in vitro assessment of its activity on human keratinocytes. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 46:e00889. [PMID: 40235517 PMCID: PMC11997404 DOI: 10.1016/j.btre.2025.e00889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/16/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025]
Abstract
Human interleukin-15 (hIL-15) is a cytokine essential for immune modulation with therapeutic applications in cancer and chronic wound healing. Although hIL-15 is commercially available, large-scale production studies remain limited. With promising clinical trial results, demand for hIL-15 is expected to rise. Plant expression systems offer a sustainable, low-cost alternative for rapid biopharmaceutical production. In this study, we optimized hIL-15 expression in Nicotiana benthamiana and assessed its physicochemical properties and biological activity. We fused hIL-15 to the Fc domain of human IgG1 for efficient purification. Through optimization of the pre- and post-infiltration conditions, we achieved transient expression and recovery at 4 dpi, yielding 33.8 µg/g fresh weight. Peptide mapping confirmed 97 % overall sequence coverage of the primary structure. Treatment with plant-produced hIL-15-Fc effectively promoted human keratinocyte HaCaT cell proliferation and migration in vitro. These findings demonstrated the potential of plant-based platforms for producing therapeutic recombinant hIL-15 that support wound healing.
Collapse
Affiliation(s)
- Chalatorn Charnsatabut
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | - Iksen Iksen
- Department of Research and development, Provenedge Co. Ltd., Bangkok 10330, Thailand
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
111
|
Singh AS, Pathak D, Jain S, Devi MS, Nongthomba U. Evaluating the potential toxicity of ampicillin using Drosophila melanogaster as a model organism. Toxicol Rep 2025; 14:101992. [PMID: 40206788 PMCID: PMC11979408 DOI: 10.1016/j.toxrep.2025.101992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
Antibiotic resistance is an indispensable threat facing in the present era. However, the studies on long term and trans-generational effects of using drugs or antibiotics on living organisms are scarce. Emphasizing the necessity to address such problems, this study investigated the potential effects of antibiotic, ampicillin (AMP) stress on the physiology of Drosophila melanogaster across multiple generations with mechanistic details. We evaluated the larval feeding behavior, fertility, cell viability in ovary and testis, longevity, expression of methylation-related genes (dDnmt2 and dMBD2/3), and antimicrobial peptide production. Larvae exposed to AMP exhibited increased mouth hook movement, indicating altered behaviour. AMP stress significantly reduced fertility across generations, with eclosion counts decreasing notably in F3 and F4 generations compared to controls. Moreover, AMP-treated flies showed decreased cell viability in ovary and testis, leading to impaired reproductive function. AMP exposure shortened the mean lifespan of flies and upregulated the expression of apoptosis-related gene p53 in females. However, there was no significant difference in p53 expression in males. Additionally, AMP stress caused a significant decrease in Drosomycin expression in treated males, while no significant changes were observed in Drosocin and Metchnikowin. In treated females, Drosocin and Drosomycin expression increased significantly, whereas the increase in Metchnikowin was not significant. The study also revealed downregulation of methylation-related genes (dDnmt2 and dMBD2/3) in AMP-treated female flies which was normalised in the rescue flies suggesting disrupted epigenetic mechanisms. Overall, the findings highlighted the importance of evaluating the trans-generational impacts of AMP stress on Drosophila physiology and gene expression, particularly in reproductive function and epigenetic regulation. The study of the impact of widely used antibiotic, AMP on model organism, Drosophila (model organism known for its genetic similarity to human), will help in predicting potential impacts on higher organisms and human. The finding would ultimately promote proper use of antibiotics and use of alternative medicine.
Collapse
Affiliation(s)
- Asem Sanjit Singh
- Developmental and Biomedical Genetics Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Dhruv Pathak
- Developmental and Biomedical Genetics Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Sakshi Jain
- Developmental and Biomedical Genetics Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | | | - Upendra Nongthomba
- Developmental and Biomedical Genetics Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru 560012, India
| |
Collapse
|
112
|
Askarizadeh F, Butler AE, Kesharwani P, Sahebkar A. Regulatory effect of curcumin on CD40:CD40L interaction and therapeutic implications. Food Chem Toxicol 2025; 200:115369. [PMID: 40043936 DOI: 10.1016/j.fct.2025.115369] [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: 11/14/2024] [Revised: 01/24/2025] [Accepted: 03/02/2025] [Indexed: 04/21/2025]
Abstract
Natural compounds have garnered significant attention as potential therapeutic agents due to their inherent properties. Their notable qualities, including safety, efficacy, favorable pharmacokinetic properties, and heightened effectiveness against certain diseases, particularly inflammatory conditions, make them particularly appealing. Among these compounds, curcumin has attracted considerable interest for its unique therapeutic properties and has therefore been extensively studied as a potential therapeutic agent for treating various diseases. Curcumin exhibits diverse anti-inflammatory, antioxidant, and antimicrobial effects. Curcumin's immune system regulatory ability has made it a promising compound for treatment of various inflammatory diseases, such as psoriasis, atherosclerosis, asthma, colitis, IBD, and arthritis. Among the signaling pathways implicated in these conditions, the CD40 receptor together with its ligand, CD40L, are recognized as central players. Studies have demonstrated that the interaction between CD40 and CD40L interaction acts as the primary mediator of the immune response in inflammatory diseases. Numerous studies have explored the impact of curcumin on the CD40:CD40L pathway, highlighting its regulatory effects on this inflammatory pathway and its potential therapeutic use in related inflammatory conditions. In this review, we will consider the evidence concerning curcumin's modulatory effects in inflammatory disease and its potential therapeutic role in regulating the CD40:CD40L pathway.
Collapse
Affiliation(s)
- Fatemeh Askarizadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
113
|
Zhang W, Kong D, Zhang X, Hu L, Nian Y, Shen Z. T cell aging and exhaustion: Mechanisms and clinical implications. Clin Immunol 2025; 275:110486. [PMID: 40120658 DOI: 10.1016/j.clim.2025.110486] [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: 01/25/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
T cell senescence and exhaustion represent critical aspects of adaptive immune system dysfunction, with profound implications for health and the development of disease prevention and therapeutic strategies. These processes, though distinct, are interconnected at the molecular level, leading to impaired effector functions and reduced proliferative capacity of T cells. Such impairments increase susceptibility to diseases and diminish the efficacy of vaccines and treatments. Importantly, T cell senescence and exhaustion can dynamically influence each other, particularly in the context of chronic diseases. A deeper understanding of the molecular mechanisms underlying T cell senescence and exhaustion, as well as their interplay, is essential for elucidating the pathogenesis of related diseases and restoring dysfunctional immune responses. This knowledge will pave the way for the development of targeted therapeutic interventions and strategies to enhance immune competence. This review aims to summarize the characteristics, mechanisms, and disease associations of T cell senescence and exhaustion, while also delineating the distinctions and intersections between these two states to enhance our comprehension.
Collapse
Affiliation(s)
- Weiqi Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Dejun Kong
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Xiaohan Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Lu Hu
- Tianjin Medical University First Central Clinical College, Tianjin, China.
| | - Yeqi Nian
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China; Department of Kidney Transplant, Tianjin First Central Hospital, Tianjin, China.
| | - Zhongyang Shen
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China.
| |
Collapse
|
114
|
Tian Y, Zhang J, Jia Z, Pan X, Hu Z, Kang R, Zhou X, Luo L, Shen Z, Shen Q. Biomimetic mineralized mesenchymal stem cell-derived exosomes for dual modulation of ferroptosis and lactic acid-driven inflammation in acute liver injury therapy. J Colloid Interface Sci 2025; 687:489-506. [PMID: 39970589 DOI: 10.1016/j.jcis.2025.02.078] [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: 12/11/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Acute liver injury (ALI) is characterized by rapid and severe hepatocellular damage, leading to ferroptosis and an exacerbated inflammatory response. Mesenchymal stem cell-derived exosomes (MSC-exo) have emerged as a promising therapeutic strategy for ALI due to their ability to deliver antioxidants and stabilize solute carrier family 7 members 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) system. In this study, we developed a novel engineered exosome, MSC-exo/MnO2@DEX, by encapsulating the anti-inflammatory drug dexamethasone (DEX) within MSC-exo and modifying its surface with manganese dioxide (MnO2) via a bionano-mineralization approach. MnO2 exhibits multi-enzymatic activity, enabling efficient scavenging of reactive oxygen species (ROS), such as hydrogen peroxide and superoxide anions. When combined with MSC-exo, MnO2 not only reduces ROS levels and generates oxygen but also stabilizes the SLC7A11/GPX4 axis, thereby protecting hepatocytes from ferroptosis. Concurrently, DEX suppresses the nuclear factor-κB (NF-κB) signaling pathway, inhibits macrophage M1 polarization, and alleviates hepatic inflammation. The oxygen produced by MnO2 catalysis further mitigates hypoxia, decreases lactic acid accumulation, and downregulates histone lactylation, synergizing with DEX to enhance NF-κB pathway inhibition and amplify anti-inflammatory effects. Transcriptomic analyses revealed that MSC-exo/MnO2@DEX significantly enhances antioxidant capacity, metabolic processes, and immune function, while improving liver function and suppressing ferroptosis, lactylation and inflammatory responses. Collectively, these findings demonstrate the therapeutic potential of MSC-exo/MnO2@DEX as an effective treatment for ALI.
Collapse
Affiliation(s)
- Yiwei Tian
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jun Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zengguang Jia
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiuhua Pan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zongwei Hu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ruixin Kang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiawei Zhou
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lin Luo
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ziqi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
115
|
Xia J, Wang Y, Li X, Liu L, Zhang P, Dai W, Luo P, Wang G, Li Y. The mechanism of perilla oil in regulating lipid metabolism. Food Chem 2025; 476:143318. [PMID: 39977980 DOI: 10.1016/j.foodchem.2025.143318] [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: 09/18/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025]
Abstract
Emerging science supports the role of lipid metabolism disorders in the occurrence and development of chronic diseases. Dietary intervention has been shown to be an effective strategy for regulating lipid metabolism. Recent studies showed that perilla is rich in various effective ingredients, including fatty acids, flavonoids, and phenolic acids. These ingredients exhibit a myriad of benefits, notably enhancing intestinal health and helping to manage metabolic diseases. Perilla oil stands out as a promising agent for regulating lipid metabolism, underscoring its potential for various health applications. This review introduces the active ingredients in perilla and provides a systematic overview of the mechanism by which perilla oil regulates lipid metabolism to expand its application value. Further research should focus on exploring the dose effect and absorption efficiency of perilla oil in clinical applications.
Collapse
Affiliation(s)
- Jiawei Xia
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Yi Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Xin Li
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Li Liu
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Pin Zhang
- Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Wendong Dai
- Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Peng Luo
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Guoze Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| | - Yanhong Li
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
116
|
Liu F, Howard CB, Huda P, Fletcher NL, Bell CA, Blakey I, Agrez M, Thurecht KJ. Immune-modulating nanomedicines for enhanced drug delivery to non-small-cell lung cancer. Biomaterials 2025; 317:123089. [PMID: 39793167 DOI: 10.1016/j.biomaterials.2025.123089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Immune-modulating peptides have shown potential as novel immune-stimulating agents which enhance the secretion of anticancer cytokines in vitro. However, fast clearance from blood hampers the ability of such peptides to accumulate in the tumour and results in limited therapeutic efficacy in animal studies. To address the fast blood clearance, this work reports the development and validation of a novel polymeric nanoparticle delivery system for the efficient localization of an immunomodulating peptide in the tumour microenvironment (TME). To identify the optimal polymeric nanoparticle for this study, two types of nanoparticles were developed as either branched polymers or micelles that have similar chemical functionality but different sizes. The effect of targeting the nanomedicine to the tumour-specific antigen, glycoprotein GPC-1, was explored using a bispecific antibody (BsAb) that shows an affinity for the cell protein (GPC-1) and the nanoparticle. These systems were evaluated for targeting efficiency and tumour penetration using tumour spheroids of Lewis Lung Cancer (LLC) cells and it was shown that the targeted system significantly enhanced cell association compared to the untargeted control with minor differences in penetration. The lead micelle-peptide conjugates were identified and using in vivo allograft models they were demonstrated to have high delivery efficiency of the peptide to tumours, prolonged blood circulation, enhanced tumour accumulation and tumour suppression that was associated with immune cell recruitment to the tumour.
Collapse
Affiliation(s)
- Feifei Liu
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Pie Huda
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Craig A Bell
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Idriss Blakey
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael Agrez
- ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia; InterK Peptide Therapeutics Limited, Sydney, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia.
| |
Collapse
|
117
|
Joshi G, Décembre E, Brocard J, Montpellier C, Ferrié M, Allatif O, Mehnert AK, Pons J, Galiana D, Dao Thi VL, Jouvenet N, Cocquerel L, Dreux M. Plasmacytoid dendritic cell sensing of hepatitis E virus is shaped by both viral and host factors. Life Sci Alliance 2025; 8:e202503256. [PMID: 40175091 PMCID: PMC11966012 DOI: 10.26508/lsa.202503256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Type I and III interferons critically protect the host against viral infection. Previous studies showed that IFN responses are suppressed in cells infected by hepatitis E virus (HEV). Here, we studied the anti-HEV function of IFN secreted by plasmacytoid dendritic cells (pDCs), specialized producers of IFNs. We showed that pDCs co-cultured with HEV-replicating cells secreted IFN in a cell contact-dependent manner. This pDC response required the endosomal nucleic acid sensor TLR7 and adhesion molecules. IFNs secreted by pDCs reduced viral spread. Intriguingly, ORF2, the capsid protein of HEV, can be produced in various forms by the infected cells, and we wanted to study their role in anti-HEV immune response. During infection, a fraction of ORF2 localizes into the nucleus, and glycosylated forms of ORF2 are massively secreted by infected cells. We showed that glycosylated ORF2 potentiates the recognition of infected cells by pDCs, by regulating cell contacts. On the other hand, nuclear ORF2 triggers immune response by IRF3 activation. Together, our results suggest that pDCs may be essential to control HEV replication.
Collapse
Affiliation(s)
- Garima Joshi
- CIRI, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University Lyon, Lyon, France
| | - Elodie Décembre
- CIRI, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University Lyon, Lyon, France
| | - Jacques Brocard
- Université Claude Bernard Lyon 1, CNRS UAR3444, INSERMUS8, ENS de Lyon, SFR Biosciences, Lyon, France
| | - Claire Montpellier
- University Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Martin Ferrié
- University Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Omran Allatif
- CIRI, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University Lyon, Lyon, France
| | - Ann-Kathrin Mehnert
- Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany and German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Johann Pons
- Sup'biotech, École Des Ingénieurs En Biotechnologies, Villejuif, Paris
| | - Delphine Galiana
- CIRI, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University Lyon, Lyon, France
| | - Viet Loan Dao Thi
- Department of Infectious Diseases, Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany and German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Nolwenn Jouvenet
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Virus sensing and signaling Unit, Paris, France
| | - Laurence Cocquerel
- University Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Lille, France
| | - Marlène Dreux
- CIRI, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, University Lyon, Lyon, France
| |
Collapse
|
118
|
Ma L, Wang W, Gu L, Wang L. cPLA 2α on the influence of Th17 and its role in the formation of liver fibrosis. Cytotechnology 2025; 77:87. [PMID: 40206205 PMCID: PMC11977053 DOI: 10.1007/s10616-025-00750-6] [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: 01/07/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
This study primarily investigated the mechanism and pathways of the cPLA2α signaling pathway on Th17-mediated HSC activation and liver fibrosis, providing insights for clinical strategies to target HSC activation and delay the rapid progression of liver fibrosis. In vitro and in vivo model were established, and different concentrations of the cPLA2α inhibitor AACOCF3 were administered respectively for intervention. The expression of IL- 17 was detected by ELISA, and the expression of cPLA2α protein and HSC activation protein α-SMA index were detected by Western blot and immunofluorescence. In addition, observe the changes in the degree of liver fibrosis in mice through the pathological staining of mouse livers. In an in vitro system, Th17 could induce HSC activation. And after intervention, the results showed that the inhibitor could inhibit Th17 activation of HSC. Next, in an in vivo model, Th17 could also induce HSC activation. And after intervention, the results showed that the inhibitor could also inhibit HSC activation by Th17. Observation under liver pathological staining showed that the inflammation and staining were significantly reduced in the intervention group, suggesting a therapeutic effect of AACOCF3. Using in vitro and in vivo approaches, these data suggest that Th17 cells can promote the activation and proliferation of HSCs, which further exerts a role in promoting liver fibrosis. These data also suggest that the cPLA2α pathway may be involved in the activation of HSCs by Th17 cells and induce liver fibrosis mechanisms.
Collapse
Affiliation(s)
- Lina Ma
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
- Shandong First Medical University, Jinan, 250117 Shandong China
| | - Wei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
- Shandong First Medical University, Jinan, 250117 Shandong China
| | - Limin Gu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
- Shandong First Medical University, Jinan, 250117 Shandong China
| | - Liyun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 Shandong China
| |
Collapse
|
119
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [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: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
120
|
Fan G, Liu Y, Tao L, Wang D, Huang Y, Yang X. Sodium butyrate alleviates colitis by inhibiting mitochondrial ROS mediated macrophage pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167756. [PMID: 40044062 DOI: 10.1016/j.bbadis.2025.167756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with unclear causes and limited treatment options. Sodium butyrate (NaB), a byproduct of dietary fiber in the intestine, has demonstrated efficacy in treating inflammation. However, the precise anti-inflammatory mechanisms of NaB in colon inflammation remain largely unexplored. This study aims to investigate the effects of NaB on dextran sulfate sodium (DSS)-induced colitis in rats. The findings indicate that oral administration of NaB effectively prevent colitis and reduce levels of serum or colon inflammatory factors. Additionally, NaB demonstrated in vitro inhibition of RAW264.7 inflammation cytokines induced by LPS, along with suppression of the ERK and NF-κB signaling pathway activation. Moreover, NaB mitigated LPS and Nigericin-induced RAW264.7 pyroptosis by reducing indicators of mitochondrial damage, including increased mitochondrial membrane potential (JC-1) levels and decreased Mito-ROS production. NaB increases ZO-1 and Occludin expression in CaCo2 cells by inhibiting RAW264.7 pyroptosis. These results suggest that NaB could be utilized as a therapeutic agent or dietary supplement to alleviate colitis.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaxin Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Limei Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Danping Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yizhu Huang
- Singao Xiamen Company, Xiamen 361006, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
121
|
Tiwari A, Khanam A, Mandal PK. Organocatalyzed O-glycosylation of glycosyl trichloroacetimidates donors: l-prolinethioamide as brønsted acid catalyst. Carbohydr Res 2025; 552:109470. [PMID: 40174324 DOI: 10.1016/j.carres.2025.109470] [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: 12/20/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
In this study, we present the utilization of l-proline-derived thioamide small organic molecules as an effective organocatalyst for the O-glycosylation of various glycosyl trichloroacetimidate donors, eliminating the need for any cocatalysts or additives. The catalytic process achieves high yields with a wide array of alcohol and sugar nucleophiles, demonstrating a broad substrate scope and operational simplicity under mild reaction conditions. Preliminary mechanistic investigations indicate that l-prolinethioamide facilitates the glycosylation reaction via Brønsted acid/base catalysis, involving the formation of a catalyst-acceptor adduct.
Collapse
Affiliation(s)
- Ashwani Tiwari
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, P.O. Box 173, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ariza Khanam
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, P.O. Box 173, Lucknow, 226 031, India
| | - Pintu Kumar Mandal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, P.O. Box 173, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
122
|
Yang Z, Cao P, Xiao W, Song F, Wu X, Zhang X, He J, Buttino I, Yan X, Liao Z. Different molecular responses of Mytilus mantle to lipopolysaccharide and peptidoglycan challenges. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110246. [PMID: 40037495 DOI: 10.1016/j.fsi.2025.110246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Mytilus live in water as sessile filter feeders, and the mantle tissue plays an important role in their immune defense. However, the overall knowledge of the immunity of this tissue remains limited. Peptidoglycan (PGN) and lipopolysaccharide (LPS) are the most representative microbe-associated molecular patterns (MAMPs) that play roles in the immune stimulation of host cells. In the present study, ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS)-based metabolomic analysis was performed to understand the precise regulatory mechanism at the molecular level in the Mytilus mantle in response to PGN and LPS stress. Moreover, the antioxidant ability and free amino acid composition of the mantle, and the antimicrobial activities of mantle mucus were evaluated. Our results revealed that LPS and PGN stresses had different effects on the mantle's free amino acid composition and antioxidant ability, and the mantle mucus' antimicrobial activity. Both PGN and LPS stress-induced alterations in amino acids, phospholipids, fatty acids, nucleotides, and their derivatives in the mantle. PGN injection activated the amino acid-related metabolism, and inhibited the lipid-related metabolisms in the mantle, while LPS injection activated the amino acid-related metabolisms and inhibited the arachidonic acid metabolism in the mantle compared to that in the control group. In addition, activation of the mTOR and FoxO signaling pathways and inhibition of lipid-related metabolism were observed in PGN vs. LPS. In addition, PGN injection induced the upregulation of fosfomycin and deoxynojirimycin in the mantle compared to LPS injection. Our study highlights the different responses at the metabolomic level of the mussel mantle to different MAMPs and the potential application of metabolites that specifically respond to PGN and LPS challenges in mussels as biomarkers.
Collapse
Affiliation(s)
- Zilin Yang
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Pingling Cao
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Wenhui Xiao
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Fang Song
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Xiaoshan Wu
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Xiaolin Zhang
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Jianyu He
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Isabella Buttino
- Italian Institute for Environmental Protection and Research (ISPRA), Via Vitaliano Brancati 48, 00144, Rome, Italy
| | - Xiaojun Yan
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China
| | - Zhi Liao
- Laboratory of Marine Biology Protein Engineering, Marine Science and Technical College, Zhejiang Ocean University, Zhoushan City, 316022, Zhejiang, China.
| |
Collapse
|
123
|
Anghelache M, Voicu G, Anton R, Safciuc F, Boteanu D, Deleanu M, Turtoi M, Simionescu M, Manduteanu I, Calin M. Inflammation resolution-based treatment of atherosclerosis using biomimetic nanocarriers loaded with specialized pro-resolving lipid mediators. Mater Today Bio 2025; 32:101733. [PMID: 40255582 PMCID: PMC12008601 DOI: 10.1016/j.mtbio.2025.101733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Recent studies have shown that chronic inflammation in atherosclerotic (ATH) lesions is due to an inability to resolve the inflammatory response. We evaluated the therapeutic potential of specialized pro-resolving mediators (SPMs) incorporated into biomimetic lipid nanoemulsions covered with macrophage membranes (Bio-LN/SPMs) to enhance their stability, targeting, and bioactivity in resolving atherosclerotic plaque inflammation. We utilized both in vitro and in vivo experimental models to test this hypothesis. In vitro, we found that Bio-LN/SPMs significantly reduced the inflammatory markers VCAM-1, MCP-1 in TNF-α-activated endothelial and smooth muscle cells, and iNOS, and NLRP3 in LPS-activated macrophages. In contrast, free SPMs exhibited a more modest effect. In vivo, the i.v. administration of Bio-LN/SPMs in ApoE-deficient mice with progressive atherosclerotic lesions developed after administration for 4 and 8 weeks of a high-fat diet, reduced plasma triglycerides, improved renal function, and decreased plasma proteins associated with complement activation and inflammation (i.e. C4d, C5b-9, IL-6, and MCP-1) to a greater extent than other treatment groups. Bio-LN/SPMs also affected circulated monocyte subpopulations by increasing the percentage of anti-inflammatory Ly6Clow monocytes and reducing that of pro-inflammatory Ly6Chigh monocytes. Additionally, they promoted the transition of macrophages in atherosclerotic plaques to a reparative M2 phenotype. They decreased the production of TNF-α, IL-1β, and IL-6 cytokines, along with lipid deposits in the aorta of ApoE-deficient mice. These findings demonstrate the improved therapeutic efficacy of Bio-LN/SPMs compared to unincorporated SPMs and standard nanoemulsions (LN/SPMs), emphasizing their potential as a novel approach for treating atherosclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Maria Anghelache
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Geanina Voicu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Ruxandra Anton
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Florentina Safciuc
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Delia Boteanu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Mariana Deleanu
- “Liquid and Gas Chromatography” Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Mihaela Turtoi
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Maya Simionescu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Ileana Manduteanu
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| | - Manuela Calin
- "Medical and Pharmaceutical Bionanotechnologies" Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568, Bucharest, Romania
| |
Collapse
|
124
|
Giovarelli M, Mocciaro E, Carnovale C, Cervia D, Perrotta C, Clementi E. Immunosenescence in skeletal muscle: The role-play in cancer cachexia chessboard. Semin Cancer Biol 2025; 111:48-59. [PMID: 40020976 DOI: 10.1016/j.semcancer.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
With the increase in life expectancy, age-related conditions and diseases have become a widespread and relevant social burden. Among these, immunosenescence and cancer cachexia play a significant often intertwined role. Immunosenescence is the progressive aging decline of both the innate and adaptive immune systems leading to increased infection susceptibility, poor vaccination efficacy, autoimmune disease, and malignancies. Cancer cachexia affects elderly patients with cancer causing severe weight loss, muscle wasting, inflammation, and reduced response to therapies. Whereas the connections between immunosenescence and cancer cachexia have been raising attention, the molecular mechanisms still need to be completely elucidated. This review aims at providing the current knowledge about the interplay between immunosenescence, skeletal muscle, and cancer cachexia, analyzing the molecular pathways known so far to be involved. Finally, we highlight potential therapeutic strategies suited for elderly population aimed to block immunosenescence and to preserve muscle mass in cachexia, also presenting the analysis of the current state-of-the-art of related clinical trials.
Collapse
Affiliation(s)
- Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| | - Emanuele Mocciaro
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo 01100, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan 20157, Italy.
| |
Collapse
|
125
|
Hu Y, Zhang C, Wu Z, Wang S, Bao Y. Genome-wide identification and immune response analysis to vibrio for heme peroxidase in the blood clam Anadara granosa. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101430. [PMID: 39923660 DOI: 10.1016/j.cbd.2025.101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/02/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Heme peroxidases (HPs), an essential class of oxidoreductases that may have an important role in molluscan immune defense, remain inadequately studied in mollusks. In this study, HPs in Anadara granosa (AgHPs) were comprehensively analyzed on a genome-wide scale and their immune responses under stress from three species of Vibrio were investigated. A total of 18 AgHPs were identified in the genome of A. granosa, unequally distributed in four subfamilies, peroxinectin (Pxt), prostaglandin G/H synthase (PGHS), dual oxidases (DuOx) and peroxidasin (Pxd). AgHPs were mainly expressed in gills and mantle, with AgHP6 being the only member expressed in hemocytes. The immune response of A. granosa to Vibrio stress demonstrated dynamic alterations in gene expression, characterized by prominent upregulation of AgHP3 and AgHP4 in both gills and mantle, alongside significant downregulation of several other genes; these expression changes likely underpin antimicrobial immunity in A. granosa and suggest distinct regulatory mechanisms and functional roles. Further analyses suggest that AgHP11 (Pxt) may enhance cell adhesion when invaded by exogenous bacteria, and AgHP3 (PGHS) and AgHP4 (PGHS) contribute to the immunity by synthesizing various prostaglandins. AgHP6 (DuOx) and AgHP7 (DuOx) may activate epithelial immunity by reducing expression levels. Additionally, during the anti-Vibrio process, hemoglobin may substitute for ROS-producing HPs to provide antimicrobial defense in A. granosa. This study represents the first genome-wide identification and immune response analysis of HPs in A. granosa, laying a foundation for future research into the immune functions of HPs in mollusks.
Collapse
Affiliation(s)
- Yazhuo Hu
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang 315100, China
| | - Chengtian Zhang
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang 315100, China
| | - Zongming Wu
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang 315100, China
| | - Sufang Wang
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang 315100, China; Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai 315604, China.
| | - Yongbo Bao
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang 315100, China; Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai 315604, China.
| |
Collapse
|
126
|
Wei J, Qin Y, Zhang L, Gong X. Combined protein and transcriptomics identifies DCTPP1 as a putative biomarkers for predicting immunotherapy responsiveness in gastric cancer patients. Anticancer Drugs 2025; 36:415-426. [PMID: 39908235 DOI: 10.1097/cad.0000000000001704] [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] [Indexed: 02/07/2025]
Abstract
This study aimed to screen the changes after overexpression of dCTP pyrophosphatase 1 (DCTPP1) in human gastric adenocarcinoma cells (AGS) cells by proteome and transcriptome sequencing. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed to explore the functional significance of the differentially expressed DCTPP1 in gastric cancer (GC). Cell Counting Kit-8 (CCK-8) assay was used to detect the proliferation of cells. Western blot was used to detect the expression of proteins. A total of 28 genes that were significantly associated with DCTPP1 overexpression and had prognostic value were screened by Cox regression analysis. The results of gene set enrichment analysis showed that the genomes of patients with subtype A exhibited significant enrichment in pathways such as DNA repair, pyrimidine synthesis, and glucose metabolism. The tumor immune dysfunction and exclusion and The Cancer Immunome Atlas databases showed that patients with type A GC were better candidates for immunotherapy than patients with type B GC. Furthermore, the CCK-8 assay indicated significantly enhanced proliferative activity after overexpressing DCTPP1 in AGS cells, corroborating the findings from the bioinformatic analysis. The data suggest a potential association between DCTPP1 expression and both the prognosis of GC patients and the efficacy of immunotherapy. These findings offer valuable insights for the potential optimization of therapeutic strategies in gastric cancer.
Collapse
Affiliation(s)
- Jun Wei
- Department of Gastroenterology, General Practice, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical University
| | | | - Luwen Zhang
- School of Clinic Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Xiaobing Gong
- Department of Gastroenterology, General Practice, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province
| |
Collapse
|
127
|
Kazemi M, Sanati M, Shekari Khaniani M, Ghafouri-Fard S. A review on the lncRNA-miRNA-mRNA regulatory networks involved in inflammatory processes in Alzheimer's disease. Brain Res 2025; 1856:149595. [PMID: 40132722 DOI: 10.1016/j.brainres.2025.149595] [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: 01/22/2025] [Revised: 03/01/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Alzheimer's disease is a progressive neurodegenerative condition that is the most frequent reason for dementia. Due to the increasing trend of aging in societies, it will place a large social and financial burden on society. Although beta amyloid plaques and the formation of neurofibrillary tangles are mentioned as the main events in this disorder, the exact molecular pathology and inflammatory regulatory networks involved in neuroinflammatory events, as a fundamental pathogenic mechanism remain unknown. Understanding these molecular network pathways in addition to helping to understand the pathogenesis of Alzheimer's disease, can also help in the early diagnosis as well as the control of inflammatory processes that are involved in its progression. So, in this study, we intend to have an overview on the regulatory lncRNAs of Alzheimer's disease and their related miRNA and mRNAs, as well as the relationship of these regulatory pathways with inflammatory processes, so that we can provide a perspective for future studies in the field of diagnosis and possibly treatment of this disorder.
Collapse
Affiliation(s)
- Masoumeh Kazemi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahla Sanati
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Shekari Khaniani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
128
|
Zeiträg J, Benedicic M, Wolf J, Ammon T, Mayr V, Holthoff HP, Kahaly GJ, Ungerer M. Inflammatory and tolerogenic dendritic cells and T lymphocytes in Graves' thyroidal and orbital disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167747. [PMID: 40024060 DOI: 10.1016/j.bbadis.2025.167747] [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: 11/18/2024] [Revised: 01/31/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Graves' disease (GD) and orbitopathy (GO) are common systemic autoimmune diseases targeting the human thyrotropin receptor. We carried out detailed immune cell investigations in GD/GO. METHODS AND RESULTS For the first time, we describe inflammatory and tolerogenic dendritic cells (tolDCs) derived from GD/GO patient monocytes. GD/GO tolDCs show reduced inflammatory marker CD86 expression, but higher CD11c levels. CD86 expression on DCs correlates to anti-TSHR antibody titers in the patients. Release of interleukin-10 from GD/GO tolDCs is increased, and their phagocytotic capacity is maintained, whereas it is reduced in inflammatory DCs. We also characterise follicular T helper cells (Tfh) and detect significantly increased circulating Tfh17 in GD/GO. GD/GO Tfh cells were efficiently differentiated from CD4+ T cells in vitro. CONCLUSIONS Since DCs and Tfh are pivotal in B cell responses, these results are fundamental for future co-culture of disease-relevant germinal centres to model human GD/GO and test novel therapeutic approaches.
Collapse
Affiliation(s)
- Julia Zeiträg
- ISAR Bioscience Institute, Semmelweisstrasse 5, 82152 Planegg, Germany
| | - Manuela Benedicic
- ISAR Bioscience Institute, Semmelweisstrasse 5, 82152 Planegg, Germany
| | - Jan Wolf
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Tim Ammon
- ISAR Bioscience Institute, Semmelweisstrasse 5, 82152 Planegg, Germany
| | - Vanessa Mayr
- ISAR Bioscience Institute, Semmelweisstrasse 5, 82152 Planegg, Germany
| | | | - George J Kahaly
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Martin Ungerer
- ISAR Bioscience Institute, Semmelweisstrasse 5, 82152 Planegg, Germany.
| |
Collapse
|
129
|
Zhang H, Jiang N, Xu M, Jing D, Dong T, Liu Q, Lv Q, Huo R, Chen P, Li L, Wang X. M2 macrophage derived exosomal miR-20a-5p ameliorates trophoblast pyroptosis and placental injuries in obstetric antiphospholipid syndrome via the TXNIP/NLRP3 axis. Life Sci 2025; 370:123561. [PMID: 40127859 DOI: 10.1016/j.lfs.2025.123561] [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: 12/07/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
AIM Obstetric antiphospholipid syndrome (OAPS) is a pregnancy-related complication characterized by trophoblast pyroptosis and placental injury induced by antiphospholipid antibodies (aPLs). M2-polarized macrophage-derived exosomes (M2-exos) exert anti-inflammatory, immunomodulatory, and growth-promoting effects in various autoimmune diseases and tumors. However, their role in OAPS is not yet clear. Therefore, in this study, we isolated M2-exos from M2 macrophages and investigated their effects on trophoblast proliferation, death, migration, invasion, and pyroptosis following stimulation using aPLs. MAIN METHODS First, we established an animal model of OAPS and thereafter treated the OAPS mice with exogenous M2-exos via injection through the tail vein. Then to clarify the roles of miR-20a-5p and thioredoxin-interacting protein (TXNIP) in OAPS, we performed gain- or loss-of-function assays, and used GraphPad Prism software to analyze the collected data with statistical significance set at P < 0.05. KEY FINDINGS MicroRNAs (miRNAs) sequencing revealed the enrichment of miR-20a-5p in M2-exos, and these M2-exos significantly alleviated aPLs-induced trophoblast dysfunction. Our results also indicated that M2-exos delivered miR-20a-5p to trophoblast cells directly targeted thioredoxin-interacting protein (TXNIP), and thus suppressed the TXNIP/NLRP3 pathway, reduced pyroptosis and inflammation in trophoblast cells, and improved placental function and fetal development. SIGNIFICANCE M2-exos improve pregnancy outcomes in OAPS via the miR-20a-5p/TXNIP/NLRP3 axis, and thus represent as a novel therapeutic approach for aPLs-induced OAPS.
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China
| | - Ning Jiang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Mingyang Xu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Die Jing
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Tingting Dong
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Feixian County People's Hospital, Linyi 273400, Shandong, China
| | - Qingfeng Lv
- The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, Shandong, China
| | - Ruiheng Huo
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China.
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| | - Xietong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| |
Collapse
|
130
|
Xu L, Huang J, Wang R, Feng J, Wang L, Li N, Jin M, Lin H, Chen X. A novel synthetic oxazolidinone derivative BS-153 attenuated LPS-induced inflammation via inhibiting NF-κB/pkcθ signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110292. [PMID: 40122191 DOI: 10.1016/j.fsi.2025.110292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/15/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
BS-153, a new derivative of oxazolidinone, was firstly found having potent anti-inflammatory effects both in vitro and in vivo. Our study aimed to study its potential molecular mechanisms. Firstly, BS-153 significantly inhibited the expression levels of inflammatory mediators (iNOS and COX-2) and pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) on LPS-stimulated RAW264.7 cells in a dose-dependent manner. Subsequently, NF-κB nuclear translocation was blocked by 10 nM BS-153 after LPS-activated, and the phosphorylation of IκB, which could bind NF-κB and limit NF-κB nuclear translocation, was notably downregulated. The mechanistic investigation was followed the NF-κB-ikkα-TLR4/PKCθ pathway. The kinase panel screen and WB result revealed that BS-153 inhibited PKCθ phosphorylation on thr538 and ser643/676 site, and the expression of IL-17ɑ, instead of TLR4/myd88. Similarly in vivo anti-inflammatory activity was assessed by LPS-stimulation and tail-amputation in zebrafish and the results indicated that macrophages migration and infiltration were significantly inhibited by BS-153. In addition, RT-PCR results discovered that BS-153 can reduce the level of TNF-α, IL-1b and COX-2. In summary, we established BS-153 and evaluated anti-inflammatory effect for the first time. The mechanism analysis showed that BS-153 possesses anti-inflammatory activities by inhibiting the phosphorylation of PKCθ, and then leading to the inactivation of NF-κB pathway. These findings implied that BS-153 is a potential candidate for the treatment of inflammatory-related diseases.
Collapse
Affiliation(s)
- Liyan Xu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| | - Jing Huang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| | - Jinhong Feng
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250014, China
| | - Lizheng Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| | - Ning Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| | - Houwen Lin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China
| | - Xiqiang Chen
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, China
| |
Collapse
|
131
|
Liu Y, Zhao C, Liu J, Du Y. Design, synthesis, and biological evaluation of novel KRN7000 analogues using 5α-gem-difluorocarba-β-l-arabinopyranose. Carbohydr Res 2025; 552:109457. [PMID: 40081114 DOI: 10.1016/j.carres.2025.109457] [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: 01/16/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Two novel KRN7000 analogues, where d-galactopyranosyl residue was replaced by 5α-gem-difluorocarba-β-l-arabinopyranose, were designed based on docking computation and energy decomposition analyses. The target compounds were synthesized employing the key steps of Ferrier's carbocyclic ring closure and gem-difluoride formation with d-galactose as starting material. The in vivo bioassay revealed that the designed glycolipids could stimulate iNKT cells to produce cytokines IFN-γ and IL-4. The introduced hydroxyl groups on glycolipid acyl chain provided extra CD1d substrate affinities, and thus favored to boost Th1-type cytokine secretion. When the ring oxygen was replaced by CF2 group on sugar unit, its TCR affinities were enhanced in contrast with KRN7000. The in vivo cytokine profiles induced by synthetic glycolipids were initially dominated by the binding ability of CD1/glycolipid, and then adjusted by affinity toward TCR in CD1/α-GalCer/TCR triplex structure. The current results could be helpful in designing of more efficient α-GalCer analogs.
Collapse
Affiliation(s)
- Yuanfang Liu
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chuanfang Zhao
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Liu
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Binzhou, 256606, China.
| | - Yuguo Du
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Binzhou, 256606, China
| |
Collapse
|
132
|
Marunouchi T, Kyono M, Kikuchi N, Tanonaka K. Gemfibrozil mitigates caspase-11-driven myocardial pyroptosis in ischemia/reperfusion injury in mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 12:100292. [PMID: 40134584 PMCID: PMC11932663 DOI: 10.1016/j.jmccpl.2025.100292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025]
Abstract
The size of the infarct area following acute myocardial infarction (AMI) is a critical prognostic factor. Caspase-11-dependent pyroptosis has been implicated as a key mechanism driving cardiomyocyte death after AMI. However, no therapeutic agents have been developed to inhibit myocardial cell death by targeting caspase-11. This study investigates the effects of gemfibrozil, a potential caspase-11 inhibitor, on ischemia/reperfusion-induced myocardial pyroptosis in mice. To model AMI, the left coronary artery of C57BL/6 N mice was ligated for 1 h, followed by reperfusion. Levels of cleaved caspase-11 and the N-terminal fragment of gasdermin D (GSDMD-N) in ischemic myocardial tissue increased progressively over time after ischemia/reperfusion. Gemfibrozil treatment during reperfusion significantly attenuated these increases in cleaved caspase-11 and GSDMD-N levels. Moreover, gemfibrozil reduced the extent of myocardial infarct size during reperfusion. In cultured cardiomyocytes isolated from adult mice, hypoxia/reoxygenation-induced increases in caspase-11 and GSDMD cleavage were similarly mitigated by gemfibrozil, which concurrently prevented necrotic cell death. These findings demonstrate the involvement of caspase-11-dependent pyroptosis in myocardial cell death following ischemia/reperfusion and suggest that gemfibrozil holds promise as a therapeutic agent for reducing myocardial infarct size after AMI.
Collapse
Affiliation(s)
- Tetsuro Marunouchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Mayu Kyono
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Naoko Kikuchi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| | - Kouichi Tanonaka
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, Japan
| |
Collapse
|
133
|
Zhu Q, Chen Z, Wang D, Jiao X, Luan Y, Wang M, Luo R, Wang Y, Fu G, Wang Y, Zhang W. Microenvironment-responsive coating for vascular stents to regulate coagulation-inflammation interaction and promote vascular recovery. Bioact Mater 2025; 48:443-457. [PMID: 40093305 PMCID: PMC11909720 DOI: 10.1016/j.bioactmat.2025.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Early coagulation-inflammation interaction and late in-stent restenosis undermine the efficacy of vascular stents after implantation. Targeting the interplay between inflammation and coagulation, and smooth muscle cell (SMC) proliferation, we presented a microenvironment-responsive coating designed to regulate tissue responses and vascular regeneration throughout the remodeling process. Coagulation was inhibited by incorporating anticoagulant tirofiban into the coating. MMP9-responsive nanoparticles embedded in the coating released salvianolic acid A to modulate inflammatory cell behavior and inhibit SMC dysfunction. By effectively interfering with clotting and inflammation, the coating suppressed platelet-fibrin interaction and formation of platelet-monocyte aggregates, thereby mitigating adverse effects on reendothelialization. Its ability to influence SMC proliferation and migration resulted in reduced intimal hyperplasia. Coated stents were shown to significantly regulate tissue regeneration, improve the vascular environment and even reduced the lipid content in the narrowed atherosclerotic vessels in vivo. This direct approach enhanced the vascular tissue regeneration after stent implantation, and offered promising insights for optimizing vascular stent design.
Collapse
Affiliation(s)
- Qiongjun Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Zhezhe Chen
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Dan'an Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Xiaolu Jiao
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Min Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Yanan Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| |
Collapse
|
134
|
Chepy A, Collet A, Launay D, Dubucquoi S, Sobanski V. Autoantibodies in systemic sclerosis: From disease bystanders to pathogenic players. J Transl Autoimmun 2025; 10:100272. [PMID: 39917316 PMCID: PMC11799969 DOI: 10.1016/j.jtauto.2025.100272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
Autoantibodies (Aab) are recognized as key indicators in the diagnosis, classification, and monitoring of systemic autoimmune diseases (AID). Recent studies have expanded knowledge through the discovery of new antigenic targets, advanced methods for measuring Aab levels, and understanding their possible pathogenic roles in AID. This narrative review uses systemic sclerosis (SSc) as an example to highlight the importance of Aab associated with HEp-2 immunofluorescence assay positivity (traditionally referred as antinuclear antibodies [ANA]), exploring recent developments in the field. Firstly, we outline the various types of ANA found in SSc and their links with specific disease features. Newly discovered antibodies shed light on SSc cases where Aab had previously gone unidentified. Secondly, we emphasize the necessity for novel quantitative techniques to track Aab levels over time by gathering data regarding the timing of Aab occurrence relative to SSc symptoms and the relationships between Aab concentrations and disease severity. Finally, we discuss the experimental findings suggesting a potential direct role of Aab in the development of SSc. The advancements surrounding Aab provide insights into new disease mechanisms and may lead to innovative diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Aurore Collet
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
| | - Sylvain Dubucquoi
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Lille, France
| | - Vincent Sobanski
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), Lille, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
135
|
Kong L, Hu X, Xia D, Wu J, Zhao Y, Guo H, Zhang S, Qin C, Wang Y, Li L, Su Z, Zhu C, Xu S. Janus PEGylated CuS-engineered Lactobacillus casei combats biofilm infections via metabolic interference and innate immunomodulation. Biomaterials 2025; 317:123060. [PMID: 39736219 DOI: 10.1016/j.biomaterials.2024.123060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/25/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025]
Abstract
Bacterial implant-associated infections predominantly contribute to the failure of prosthesis implantation. The local biofilm microenvironment (BME), characterized by its hyperacidic condition and high hydrogen peroxide (H2O2) level, inhibits the host's immune response, thereby facilitating recurrent infections. Here, a Janus PEGylated CuS nanoparticle (CuPen) armed engineered Lactobacillus casei (L. casei) denoted as LC@CuPen, is proposed to interfere with bacterial metabolism and arouse macrophage antibiofilm function. Once LC@CuPen reached the BME, NIR irradiation-activated mild heat damages L. casei and biofilm structure. Meanwhile, the BME-responsive LC@CuPen can catalyze local H2O2 to produce toxic •OH, whereas in normal tissues, the effect of •OH production is greatly reduced due to the higher pH and lower H2O2 concentration. The released bacteriocin from damaged L. casei can destroy the bacterial membrane to enhance the penetration of •OH into damaged biofilm. Excessive •OH interferes with normal bacterial metabolism, resulting in reduced resistance of bacteria to heat stress. Finally, under the action of mild heat treatment, the bacterial biofilm lysed and died. Furthermore, the pathogen-associated molecular patterns (PAMPs) in LC@CuPen can induce M1 polarization of macrophages through NF-κB pathway and promote the release of inflammatory factors. Inflammatory factors enhance the migration of macrophages to the site of infection and phagocytose bacteria, thereby inhibiting the recurrence of infection. Generally, this engineered L. casei program presents a novel perspective for the treatment of bacterial implant-associated infections and serves as a valuable reference for future clinical applications of engineered probiotics.
Collapse
Affiliation(s)
- Lingtong Kong
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Xianli Hu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Demeng Xia
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jianghong Wu
- Department of Microbiology, College of Basic Medical Science, Department of Emergency, The Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
| | - Yangpeng Zhao
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Hua Guo
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Song Zhang
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Chun Qin
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Yanjun Wang
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Lei Li
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Zheng Su
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Chen Zhu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shuogui Xu
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
136
|
Klak K, Maciuszek M, Michalik A, Mazur M, Zawisza M, Pecio A, Nowak B, Chadzinska M. Fire in the belly: Stress and antibiotics induce dysbiosis and inflammation in the gut of common carp. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110301. [PMID: 40157582 DOI: 10.1016/j.fsi.2025.110301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Fish are exposed to numerous stressors which negatively affect their immune response and increase infection susceptibility. The risk of bacterial infections results in the excessive and preventive use of antibiotics. Therefore, we aimed to study how antibiotic treatment and restraint stress will affect the stress response, microbiota composition, gut morphology, and inflammatory reaction in common carp. Both restraint stress and antibiotic treatment increased cortisol level. Moreover, antibiotics induced dysbiosis in fish gut, manifested by a decrease in the total abundance of bacteria, and a shift in bacteria diversity, including a reduced number of Aeromonas, Bacteroides, Barnesiellaceae, Cetobacterium and Shewanella and an increased abundance of Flavobacterium. To a lesser extent, stress modified gut microbiota, as it decreased bacteria number and slightly changed the microbiota composition by decreasing Cetobacterium abundance and increasing Vibrio abundance. Microbiota of the antibiotic-treated and stressed fish shifted from the beneficial bacterial genera - Cetobacterium and Bacteroides, to the increased presence of unfavorable bacteria such as Brevinema, Flavobacterium and Desulfovibrionaceae. Stress and antibiotic-induced changes in the gut microbiota were related to the changes in the gut morphology when the higher abundance of goblet and rodlet cells and increased secretion activity of goblet cells were observed. Moreover, up-regulation of the expression of genes encoding pro-inflammatory mediators and cytokines involved in the Th17 immune response was present in the gut of the antibiotic-treated and stressed fish. We conclude that in carp antibiotics and stress alter the abundance and composition of the microbiota and induce Th17-dependent inflammatory reaction in the gut. Moreover, our results strongly suggest the interplay of the stress axis and the brain-gut-microbiota axis.
Collapse
Affiliation(s)
- Katarzyna Klak
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Magdalena Maciuszek
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Anna Michalik
- Department of Invertebrate Development and Morphology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Mikolaj Mazur
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Maria Zawisza
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Anna Pecio
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Barbara Nowak
- Institute for Marine and Antarctic Studies - Launceston, University of Tasmania, Launceston, Tasmania, Australia.
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
137
|
Xu L, Qiu J, Ren Q, Wang D, Guo A, Wang L, Hou K, Wang R, Liu Y. Gold nanoparticles modulate macrophage polarization to promote skeletal muscle regeneration. Mater Today Bio 2025; 32:101653. [PMID: 40151803 PMCID: PMC11937682 DOI: 10.1016/j.mtbio.2025.101653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
Skeletal muscle regeneration is a complex process that depends on the interplay between immune responses and muscle stem cell (MuSC) activity. Macrophages play a crucial role in this process, exhibiting distinct polarization states-M1 (pro-inflammatory) and M2 (anti-inflammatory)-that significantly affect tissue repair outcomes. Recent advancements in nanomedicine have positioned gold nanoparticles (Au NPs) as promising tools for modulating macrophage polarization and enhancing muscle regeneration. This review examines the role of Au NPs in influencing macrophage behavior, focusing on their physicochemical properties, biocompatibility, and mechanisms of action. We discuss how Au NPs can promote M2 polarization, facilitating tissue repair through modulation of cytokine production, interaction with cell surface receptors, and activation of intracellular signaling pathways. Additionally, we highlight the benefits of Au NPs on MuSC function, angiogenesis, and extracellular matrix remodeling. Despite the potential of Au NPs in skeletal muscle regeneration, challenges remain in optimizing nanoparticle design, developing targeted delivery systems, and understanding long-term effects. Future directions should focus on personalized medicine approaches and combination therapies to enhance therapeutic efficacy. Ultimately, this review emphasizes the transformative potential of Au NPs in regenerative medicine, offering hope for improved treatments for muscle injuries and diseases.
Collapse
Affiliation(s)
- Lining Xu
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Jiahuang Qiu
- Research Center of Nano Technology and Application Engineering, School of Public Health,Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Quanzhong Ren
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Dingding Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Anyi Guo
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Ling Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Radiology, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Kedong Hou
- Department of Orthopedics, Beijing Pinggu District Hospital, Beijing, 101200, China
| | - Renxian Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yajun Liu
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Spine Surgery, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| |
Collapse
|
138
|
Nair RS, Patel MN, Kannan T, Gour S, Hariharan MM, Prasanna V, Thirumalai A, Chockalingam R, Vasantharekha R, ThyagaRajan S, Priyanka HP. Effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. Toxicol In Vitro 2025; 106:106057. [PMID: 40112934 DOI: 10.1016/j.tiv.2025.106057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Estrogen signaling plays a crucial role in immune regulation and cancer metabolism, yet its impact on T-cell leukemia remains unclear. In hematological malignancies, estrogen receptor (ER) activation may influence metabolic shifts that affect cell survival and proliferation. This study investigates the in vitro effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. PURPOSE To assess how estrogen signaling influences metabolic reprogramming, inflammatory response, and survival pathways in Jurkat E6.1 cells through receptor-dependent and independent mechanisms. METHODS Jurkat E6.1 cells incubated with different concentrations of 17β-estradiol (10-12 M, 10-10 M, 10-8 M) or ER-α agonist 4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (10-10 M, 10-8 M, 10-6 M) or ER-β agonist diarylproprionitrile (10-10 M, 10-8 M, 10-6 M) with and without non-specific antagonist ICI 182,780 (10-6 M). The metabolic enzyme activities of hexokinase, pyruvate kinase, and citrate synthase were measured in cell pellets, while supernatants were analyzed for IL-6 and nitric oxide (NO) production. Additionally, PI3K/Akt pathway activation was assessed by measuring p-Akt/Total Akt expression. RESULTS A shift from glycolysis to oxidative phosphorylation was observed on treatment with 17β-estradiol with significant decline in hexokinase activity and a concomitant increase in activities of pyruvate kinase and citrate synthase. CONCLUSION 17β-estradiol mediates its effects on Jurkat E6.1 cells in vitro through receptor-subtype dependent and independent mechanisms involving metabolic enzymes (hexokinase, pyruvate kinase, citrate synthase), cytokines (IL-6), nitric oxide, and signaling molecules (p-Akt).
Collapse
Affiliation(s)
- Rahul S Nair
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Mantavya N Patel
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Thangamani Kannan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Shaili Gour
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Murali M Hariharan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Vijayarengamani Prasanna
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anupriya Thirumalai
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramanathan Chockalingam
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
139
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
140
|
Che J, Wu Y, Dong J, Jiang X, Yang L, Chen Y, Zhang J. Myelination Trajectory and Microglial Dynamics Following Repeated Sevoflurane Exposure in Developing Brain. Glia 2025; 73:1221-1240. [PMID: 39928319 DOI: 10.1002/glia.70000] [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: 04/23/2024] [Revised: 01/06/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025]
Abstract
The myelination is a critical process during brain development. This study aimed to explore the impact of volatile anesthetic sevoflurane on developing myelination and the role of microglial activation in this process. Neonatal C57BL/6J mice were exposed to sevoflurane at their postnatal 6-8 days. Neurobehavioral tests were used to assess fine motor and cognitive functions. Myelination of hippocampus (HC) and corpus callosum (CC), as well as microglial activation, were determined by western blotting and immunostaining. Lipid droplets were assessed by Oil-Red-O and Bodipy staining. Further, primary microglia were co-cultured with oligodendrocyte precursor cell (OPC) to determine the role of microglia in the proliferation and differentiation of OPC. And microglial inhibitor minocycline and CSF1R inhibitor PLX5622 were administered to assess the effects of microglial activation on developing myelination. The results showed that repeated sevoflurane exposure impaired both fine motor and cognitive functions and induced abnormal expressions of myelin-related proteins myelin basic protein (MBP) and platelet-derived growth factor α receptor (PDGFR-α). And accumulations of lipid droplets were found in the microglia of HC and CC after sevoflurane exposure. Further, the spatiotemporal response to repeated sevoflurane exposure in glial cells exhibited an aberrant myelination process and microglial polarization. The conditioned medium from sevoflurane-treated microglia inhibited the OPC proliferation and differentiation, while minocycline or PLX5622 alleviated sevoflurane-induced neuroinflammation and hypomyelination. Therefore, repeated sevoflurane exposure negatively affected OPC differentiation and myelination trajectory through hyperactivating microglia in developing brain, leading to motor and cognitive impairments, while microglial inhibition/depletion could protect against sevoflurane-induced damage on developing myelination.
Collapse
Affiliation(s)
- Ji Che
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Yuanyuan Wu
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Jing Dong
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Xuliang Jiang
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Li Yang
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Yali Chen
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| |
Collapse
|
141
|
Zhu Y, Li X, Lei X, Tang L, Wen D, Zeng B, Zhang X, Huang Z, Guo Z. The potential mechanism and clinical application value of remote ischemic conditioning in stroke. Neural Regen Res 2025; 20:1613-1627. [PMID: 38845225 PMCID: PMC11688546 DOI: 10.4103/nrr.nrr-d-23-01800] [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: 11/02/2023] [Revised: 01/30/2024] [Accepted: 03/19/2024] [Indexed: 08/07/2024] Open
Abstract
Some studies have confirmed the neuroprotective effect of remote ischemic conditioning against stroke. Although numerous animal researches have shown that the neuroprotective effect of remote ischemic conditioning may be related to neuroinflammation, cellular immunity, apoptosis, and autophagy, the exact underlying molecular mechanisms are unclear. This review summarizes the current status of different types of remote ischemic conditioning methods in animal and clinical studies and analyzes their commonalities and differences in neuroprotective mechanisms and signaling pathways. Remote ischemic conditioning has emerged as a potential therapeutic approach for improving stroke-induced brain injury owing to its simplicity, non-invasiveness, safety, and patient tolerability. Different forms of remote ischemic conditioning exhibit distinct intervention patterns, timing, and application range. Mechanistically, remote ischemic conditioning can exert neuroprotective effects by activating the Notch1/phosphatidylinositol 3-kinase/Akt signaling pathway, improving cerebral perfusion, suppressing neuroinflammation, inhibiting cell apoptosis, activating autophagy, and promoting neural regeneration. While remote ischemic conditioning has shown potential in improving stroke outcomes, its full clinical translation has not yet been achieved.
Collapse
Affiliation(s)
- Yajun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoguo Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingwei Lei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuyang Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daochen Wen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Zeng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofeng Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zichao Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zongduo Guo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
142
|
Franco-Fuquen P, Figueroa-Aguirre J, Martínez DA, Moreno-Cortes EF, Garcia-Robledo JE, Vargas-Cely F, Castro-Martínez DA, Almaini M, Castro JE. Cellular therapies in rheumatic and musculoskeletal diseases. J Transl Autoimmun 2025; 10:100264. [PMID: 39931050 PMCID: PMC11808717 DOI: 10.1016/j.jtauto.2024.100264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
A substantial proportion of patients diagnosed with rheumatologic and musculoskeletal diseases (RMDs) exhibit resistance to conventional therapies or experience recurrent symptoms. These diseases, which include autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus, are marked by the presence of autoreactive B cells that play a critical role in their pathogenesis. The persistence of these autoreactive B cells within lymphatic organs and inflamed tissues impairs the effectiveness of B-cell-depleting monoclonal antibodies like rituximab. A promising therapeutic approach involves using T cells genetically engineered to express chimeric antigen receptors (CARs) that target specific antigens. This strategy has demonstrated efficacy in treating B-cell malignancies by achieving long-term depletion of malignant and normal B cells. Preliminary data from patients with RMDs, particularly those with lupus erythematosus and dermatomyositis, suggest that CAR T-cells targeting CD19 can induce rapid and sustained depletion of circulating B cells, leading to complete clinical and serological responses in cases that were previously unresponsive to conventional therapies. This review will provide an overview of the current state of preclinical and clinical studies on the use of CAR T-cells and other cellular therapies for RMDs. Additionally, it will explore potential future applications of these innovative treatment modalities for managing patients with refractory and recurrent manifestations of these diseases.
Collapse
Affiliation(s)
- Pedro Franco-Fuquen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - David A. Martínez
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Eider F. Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan E. Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | | | - Mustafa Almaini
- Rheumatology, Allergy & Clinical Immunology Division, Mafraq Hospital, United Arab Emirates
| | - Januario E. Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
143
|
Chowdhury T, Saha A, Saha A, Chakraborty A, Das N. NeuralCodOpt: Codon optimization for the development of DNA vaccines. Comput Biol Chem 2025; 116:108377. [PMID: 39954612 DOI: 10.1016/j.compbiolchem.2025.108377] [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: 11/25/2024] [Revised: 01/08/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Inefficient gene translation, driven by organisms' codon preferences, is an emerging research area since this results in sluggish processes and diminished protein yields. Our research culminates in deriving efficient, optimized codon sequences by considering organism-specific Relative Codon Adaptiveness (RCA) ranges. In this research work, we have developed a novel algorithm, Neural Codon Optimization (NeuralCodOpt), to automate the process of codon optimization tailored to a specific organism and input sequence. Our algorithm has two main parts: the target Codon Adaptation Index generation using K-Means and the automation of sequence optimization using reinforcement learning. This algorithm has been tested across a set of 130 species, yielding highly optimal results that are quite significant compared to the previous works. NeuralCodOpt has shown a high accuracy of 86.7%, which would substantially contribute to Deoxyribonucleic Acid (DNA) vaccines by improving the efficiency of DNA expression vectors. These vectors are crucial in DNA vaccination and gene therapy as they enhance protein expression levels. By further incorporating it into plasmid construction, the translational efficiency of DNA vaccines will be significantly improved.
Collapse
Affiliation(s)
- Tapan Chowdhury
- Department of Computer Science and Engineering, Techno Main Salt Lake, EM-4/1, Sector V, Salt Lake, Kolkata, 700091, West Bengal, India.
| | - Aishwarya Saha
- Department of Computer Science and Engineering, Techno Main Salt Lake, EM-4/1, Sector V, Salt Lake, Kolkata, 700091, West Bengal, India.
| | - Ananya Saha
- Department of Computer Science and Engineering, Techno Main Salt Lake, EM-4/1, Sector V, Salt Lake, Kolkata, 700091, West Bengal, India.
| | - Arnab Chakraborty
- Department of Computer Science and Engineering, Techno Main Salt Lake, EM-4/1, Sector V, Salt Lake, Kolkata, 700091, West Bengal, India.
| | - Nibir Das
- Department of Computer Science and Engineering, Techno Main Salt Lake, EM-4/1, Sector V, Salt Lake, Kolkata, 700091, West Bengal, India.
| |
Collapse
|
144
|
Wan F, Zhuang K, Li Z, Wang X, Li W, Hou Y, You W, Jiang Y, Wang M, Zhu P. Electroacupuncture mitigates cognitive impairments in chronic hypoxia-induced mice by modulating neuroinflammation. IBRO Neurosci Rep 2025; 18:432-442. [PMID: 40144798 PMCID: PMC11937942 DOI: 10.1016/j.ibneur.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
This study investigates the therapeutic effects and mechanisms of electroacupuncture (EA) on cognitive impairment induced by chronic hypoxia (CH) in mice. Chronic hypoxia was simulated by exposing mice to a 10 % oxygen environment for 8 hours daily over 3 months. The cognitive functions of the mice were assessed through behavioral tests, including the open field test (OFT), Y-maze, and Morris water maze (MWM). Results showed that CH induced significant anxiety-like behaviors and memory impairments in mice. EA treatment, targeting the Baihui (GV20), Shenting (GV24), and Zusanli (ST36) acupoints, significantly ameliorated these behavioral deficits. Histological analyses using HE staining, Nissl staining, and TUNEL assays demonstrated that EA reduced neuronal damage, apoptosis, and myelin loss in the cerebral cortex and hippocampus. Additionally, EA treatment significantly lowered the expression of the pro-inflammatory cytokine TNF-α in brain tissues, suggesting its anti-inflammatory effects. Immunofluorescence and Western blot analyses revealed that EA inhibited the overactivation of microglia and astrocytes in the CH model. Specifically, EA suppressed the expression of Iba1 and GFAP, markers of microglial and astrocytic activation, respectively. Furthermore, EA promoted the polarization of microglia towards the M2 anti-inflammatory phenotype by downregulating iNOS and upregulating Arg1. Similarly, EA reduced the expression of C3, a marker of A1 astrocytes, thereby preventing astrocytic polarization towards the pro-inflammatory state. Organotypic brain slice cultures subjected to oxygen-glucose deprivation (OGD) confirmed that electrical stimulation, akin to EA, inhibited the activation of microglia and astrocytes under hypoxic conditions. In conclusion, EA improves cognitive function in CH-induced mice by reducing neuroinflammation, inhibiting glial cell overactivation, and promoting anti-inflammatory phenotypes. These findings highlight EA's potential as a therapeutic intervention for cognitive impairments related to chronic hypoxia.
Collapse
Affiliation(s)
- Fang Wan
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Kun Zhuang
- Dongguan Integrated Traditional Chinese and Western Medicine Hospital, Dongguan 523819, China
| | - Ziyu Li
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xiaoqing Wang
- Chinese Medicine Research Institute, Chengde Medical University, Chengde 067000, China
| | - Wenyan Li
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yunlong Hou
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang Yiling Pharmaceutical Co. Ltd., Shijiazhuang 050000, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral VesselCollateral Disease), Shijiazhuang 050000, China
| | - Wanhui You
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Yibing Jiang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Mingye Wang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Pengyu Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
145
|
Liu S, He Y, Jin L, Shi S, Zhang J, Xie W, Yang M, Zhang Q, Kong H. H3K18 lactylation-mediated SIX1 upregulation contributes to silica-induced epithelial-mesenchymal transition in airway epithelial cells. Toxicology 2025; 514:154109. [PMID: 40049282 DOI: 10.1016/j.tox.2025.154109] [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: 01/09/2025] [Revised: 03/02/2025] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Silica exposure-induced airway epithelial-mesenchymal transition (EMT) is a critical pathological process in pulmonary fibrosis. This study investigated the role of NLRP3 inflammasome, glycolysis, and histone lactylation in silica-induced EMT of human bronchial epithelial cells (16HBE). Silica exposure activated NLRP3 inflammasome, enhanced glycolysis and H3K18 lactylation, as well as induced EMT in 16HBE cells. Selective inhibition of NLRP3 inflammasome with MCC950, blockade of the interleukin 1 (IL-1) receptor with AF12198, or suppression of lactate production with oxamate effectively reduced glycolysis-mediated histone lactylation and mitigated silica-induced EMT. Moreover, silica-induced upregulation of PFKFB3, a key enzyme of glycolysis, was significantly mitigated by MCC950 or AF12198. Cut&Tag analysis revealed silica treatment led to H3K18 lactylation enrichment at transcription start sites (TSS), particularly within the promoter region of the sine oculis homeobox 1 (SIX1), which enhanced transcription of SIX1, a key transcription factor involved in EMT. Consistently, inhibition of histone lactylation by the histone acetyltransferase P300 inhibitor A-485 suppressed silica-induced SIX1 expression and EMT. These findings indicate that silica activates NLRP3 inflammasome and promotes interleukin 1β (IL-1β) production, thereafter enhancing PFKFB3-mediated glycolysis by IL-1 receptor. Lactate accumulation by glycolysis enhances H3K18 lactylation at the TSS facilitating expression of SIX1. Together, this inflammation-glycolysis-lactylation cascade involved in EMT provides new insights into the molecular mechanisms underlying silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Songtao Liu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yiting He
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Linling Jin
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Shuangshuang Shi
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Jiayi Zhang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Weiping Xie
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Mingxia Yang
- Department of Pulmonary & Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, China
| | - Qun Zhang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| | - Hui Kong
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| |
Collapse
|
146
|
Hamdy H, Aly WA, Elkord E. Investigating the functional role of BUB1B in aflatoxin B1-associated hepatocarcinogenesis. Toxicology 2025; 514:154127. [PMID: 40147685 DOI: 10.1016/j.tox.2025.154127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, stemming from a complex interplay of genetic, environmental, and lifestyle factors. Aflatoxin B1 (AFB1), a prevalent food contaminant, is a known HCC risk factor, but its molecular mechanisms remain incompletely understood. This study investigated the contribution of BUB1B, a crucial spindle assembly checkpoint regulator, in AFB1-induced hepatocyte malignant transformation, we assessed AFB1's impact on cell proliferation, viability, cell cycle regulation, and BUB1B expression. BUB1B knockdown via siRNA revealed its role in epithelial-mesenchymal transition (EMT), cell motility, and proliferation. AFB1 exposure significantly altered cell proliferation and cell cycle dynamics, correlating with increased BUB1B expression. Furthermore, we identified a significant interaction between BUB1B and the IL12A-JAK2/STAT4 signaling pathway, suggesting a mechanism for immune evasion and tumor progression. These findings highlight BUB1B's critical role in AFB1-induced hepatocarcinogenesis and establish its potential target for HCC. Further research is needed to fully elucidate the underlying molecular mechanisms and explore the therapeutic implications of BUB1B inhibition in HCC treatment.
Collapse
Affiliation(s)
- Hayam Hamdy
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Medicine, Yunnan University, Kunming, China; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, New Valley University, New Valley, Egypt
| | - Wafaa A Aly
- Department of Environmental Health, Institute of Environmental Studies, Arish University, Egypt
| | - Eyad Elkord
- Department of Biosciences and Bioinformatics & Suzhou Municipal Key Lab of Biomedical Sciences and Translational Immunology, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates; Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, UK.
| |
Collapse
|
147
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [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: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
148
|
Jomova K, Alomar SY, Valko R, Liska J, Nepovimova E, Kuca K, Valko M. Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem Biol Interact 2025; 413:111489. [PMID: 40147618 DOI: 10.1016/j.cbi.2025.111489] [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: 06/02/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Oxidative stress and chronic inflammation are important drivers in the pathogenesis and progression of many chronic diseases, such as cancers of the breast, kidney, lung, and others, autoimmune diseases (rheumatoid arthritis), cardiovascular diseases (hypertension, atherosclerosis, arrhythmia), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease), mental disorders (depression, schizophrenia, bipolar disorder), gastrointestinal disorders (inflammatory bowel disease, colorectal cancer), and other disorders. With the increasing demand for less toxic and more tolerable therapies, flavonoids have the potential to effectively modulate the responsiveness to conventional therapy and radiotherapy. Flavonoids are polyphenolic compounds found in fruits, vegetables, grains, and plant-derived beverages. Six of the twelve structurally different flavonoid subgroups are of dietary significance and include anthocyanidins (e.g. pelargonidin, cyanidin), flavan-3-ols (e.g. epicatechin, epigallocatechin), flavonols (e.g. quercetin, kaempferol), flavones (e.g. luteolin, baicalein), flavanones (e.g. hesperetin, naringenin), and isoflavones (daidzein, genistein). The health benefits of flavonoids are related to their structural characteristics, such as the number and position of hydroxyl groups and the presence of C2C3 double bonds, which predetermine their ability to chelate metal ions, terminate ROS (e.g. hydroxyl radicals formed by the Fenton reaction), and interact with biological targets to trigger a biological response. Based on these structural characteristics, flavonoids can exert both antioxidant or prooxidant properties, modulate the activity of ROS-scavenging enzymes and the expression and activation of proinflammatory cytokines (e.g., interleukin-1beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), induce apoptosis and autophagy, and target key signaling pathways, such as the nuclear factor erythroid 2-related factor 2 (Nrf2) and Bcl-2 family of proteins. This review aims to briefly discuss the mutually interconnected aspects of oxidative and inflammatory mechanisms, such as lipid peroxidation, protein oxidation, DNA damage, and the mechanism and resolution of inflammation. The major part of this article discusses the role of flavonoids in alleviating oxidative stress and inflammation, two common components of many human diseases. The results of epidemiological studies on flavonoids are also presented.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Richard Valko
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Jan Liska
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, 811 08, Bratislava, Slovakia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, 5005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
149
|
Zhang Y, Guan Y, Dai M, Yang Y, Yang F. Microcystin-LR induces lung injury in mice through the NF-κB/NLRP3 pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:385-394. [PMID: 39773316 DOI: 10.1080/15287394.2024.2443525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Microcystin-LR (MC-LR) a cyclic toxin produced by cyanobacterial species is known to exert detrimental effects on various organs, including lung. Several investigators demonstrated that MC-LR exerts pulmonary toxicity, but the underlying mechanisms remain unclear. This study aimed to investigate whether exposure to MC-LR-induced lung inflammation and examine the underlying mechanisms. Thirty specific pathogen-free (SPF) male mice were allocated into control and MC-LR treatment groups. Mice were intraperitoneally injected with physiological saline or MC-LR (20 μg/kg) daily for a total of 21 days. Our findings indicated that exposure to MC-LR-produced histopathological changes in lung tissue, including thickening of alveolar walls and inflammatory infiltration. MC-LR was found to upregulate mRNA expression levels of pro-inflammatory cytokines TNFα, IL-6, IL-1β, and IL-18. Further, MC-LR significantly elevated the expression levels of proteins associated with the NF-κB/NLRP3 pathway p-NF-κB, NLRP3, Caspase-1, ASC. The activation of NF-κB/NLRP3 pathway further promoted the release of inflammatory cytokine IL-1β and cleavage of pyroptosis-associated GSDMD protein. These findings indicate that MC-LR may induce lung inflammation by promoting cell pyroptosis via the activation of the NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Yin Zhang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying Guan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Manni Dai
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yue Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
150
|
Sun Y, Gao SQ, Wang X, Li T, Han YL, Miao SH, Zhao R, Zheng XB, Qiu JY, Jin WX, Gao CC, Zhou ML. Galectin-3 activates microglia and promotes neurological impairment via NLRP3/pyroptosis pathway following traumatic brain injury. Brain Res 2025; 1855:149560. [PMID: 40074166 DOI: 10.1016/j.brainres.2025.149560] [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: 01/17/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Externally caused traumatic brain injury (TBI) poses a woeful worldwide health concern, bringing about disability, death, and prolonged neurological impairment. Increased galectin-3 levels have been linked to unfavorable outcomes in several neurological conditions. This study explores the role of galectin-3 in TBI, specifically examining its contribution to neuroinflammation. METHODS BV2 microglia cells treated with lipopolysaccharide (LPS) and a mouse model of TBI were applied to investigate the impact of galectin-3 on neuroinflammation following TBI. Western blotting and immunofluorescence labeling were applied for evaluating protein levels and colocalization. Adeno-associated virus (AAV) that targets microglia was used to knock down galectin-3 in microglia. Nissl staining and the modified neurologic severity score were employed in evaluating neural survival and neurological function, and the cognitive impairment following TBI was assessed by the Y-Maze and Morri water maze test. RESULTS Galectin-3 expression was shown to rise dramatically after TBI, peaking between days five and seven. In vitro, BV2 cells treated with LPS showed reduced NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome activation when galectin-3 was inhibited. In LPS-activated microglia, galectin-3 inhibition specifically decreased the expression of Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), p-NF-κB, NLRP3, Apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, and Gasdermin D (GSDMD). Injection with AAV containing siRNA to knock down galectin-3 in microglia was operated on mice in vivo. Following TBI, this knockdown led to reduced NLRP3 inflammasome activation, neuronal death, neurological impairments and cognitive impairment. CONCLUSIONS Our foundings indicate that modulating microglia-derived galectin-3 following TBI to reduce neuroinflammation could serve as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Sheng-Qing Gao
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xue Wang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Li
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yan-Ling Han
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shu-Hao Miao
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ran Zhao
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiao-Bo Zheng
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing. China
| | - Jia-Yin Qiu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wang-Xuan Jin
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao-Chao Gao
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Meng-Liang Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|