51
|
Oda H, Annibaldi A, Kastner DL, Aksentijevich I. Genetic Regulation of Cell Death: Insights from Autoinflammatory Diseases. Annu Rev Immunol 2025; 43:313-342. [PMID: 40279314 DOI: 10.1146/annurev-immunol-090222-105848] [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: 04/27/2025]
Abstract
Metazoans have evolved innate antimicrobial defenses that promote cellular survival and proliferation. Countering the inevitable molecular mechanisms by which microbes sabotage these pathways, multicellular organisms rely on an alternative, perhaps more ancient, strategy that is the immune equivalent of suicide bombing: Infection triggers cell death programs that summon localized or even systemic inflammation. The study of human genetics has now unveiled a level of complexity that refutes the naive view that cell death is merely a blunt instrument or an evolutionary afterthought. To the contrary, findings from patients with rare diseases teach us that cell death-induced inflammation is a sophisticated, tightly choreographed process. We herein review the emerging body of evidence describing a group of illnesses-inborn errors of cell death, which define many of the molecular building blocks and regulatory elements controlling cell death-induced inflammation in humans-and provide a possible road map to countering this process across the spectrum of rare and common illnesses.
Collapse
Affiliation(s)
- Hirotsugu Oda
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany;
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Daniel L Kastner
- National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, Maryland, USA;
| | - Ivona Aksentijevich
- National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, Maryland, USA;
| |
Collapse
|
52
|
Cui S, Xiong W, Zhao Z, Han Y, Cui T, Qu Z, Li Z, Zhang X. Negative impact of maternal depressive symptoms on infancy neurodevelopment: a moderated mediation effect of maternal inflammation. Eur Child Adolesc Psychiatry 2025; 34:1403-1414. [PMID: 39190153 DOI: 10.1007/s00787-024-02572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Maternal depression promotes maternal inflammation and the risk of neurodevelopmental disorder in offspring, but the role of inflammation on the association between depression and neurodevelopment in offspring has not been extensively studied in humans. This study aims to examine the mediating role of maternal inflammation on the relationship between maternal depression and neurodevelopment in infants. 146 mother-child pairs were identified from Tianjin Maternal and Child Health Education and Service Cohort (Tianjin MCHESC). Maternal depression was investigated by the Center for Epidemiologic Studies Depression Scale and the Edinburgh Postnatal Depression Scale, and depressive trajectories were identified by latent class growth analysis. Inflammatory biomarkers in the three trimesters were assessed with enzyme-linked immunoassay. The Children Neuropsychological and Behavior Scale-Revision 2016 was used to measure neurodevelopment in infants. Principal component analysis was performed to identify inflammatory condition. Stepwise multiple linear regression analysis and mediation analysis were used to identify association among maternal depression, maternal inflammation and neurodevelopment in infants. Offspring in the low and moderate maternal depression groups exhibited higher adaptive behavior development quotient than those in the high maternal depression group. Higher maternal c-reactive protein level and higher inflammatory level in acute-phase of inflammation in the first trimester, and moderate maternal depression were associated with lower adaptive behavior quotients of infants. Inflammatory level in acute-phase of inflammation in the first trimester significantly mediated the association between maternal depression and adaptive behavior development of infants, with explaining 11.85% of the association. Maternal depression could impair adaptive behavior development in infants by inflammation.
Collapse
Affiliation(s)
- Shanshan Cui
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Wenjuan Xiong
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Ziyu Zhao
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Yu Han
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Tingkai Cui
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Zhiyi Qu
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Zhi Li
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Xin Zhang
- Department of Maternal, Child & Adolescence Health, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
53
|
Deng S, Zhong D, Dong Y, Qian Y, Wang B, Hu M, Liu M, Tan K, Zhang C, Tang H. Network Pharmacology and Experimental Validation Reveal Ganodermanontriol Modulates Pneumonia via TNF/NF-κB/MAPKs Signaling Pathway. Food Sci Nutr 2025; 13:e70123. [PMID: 40144560 PMCID: PMC11936839 DOI: 10.1002/fsn3.70123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Ganoderma lucidum (Leyss. ex Fr.) Karst, commonly known as Lingzhi, has long been employed in traditional Chinese medicine for its medicinal properties, particularly in alleviating respiratory issues like cough and asthma. Recognized both as a therapeutic agent and an edible supplement, Lingzhi is celebrated for its health-promoting benefits. Despite its widespread use, the effectiveness of G. lucidum in treating pneumonia has not been extensively studied, highlighting the need for further research. This research aimed to evaluate the potential of G. lucidum in pneumonia treatment and to uncover the mechanisms behind its effects, specifically examining how its active constituents influence inflammatory pathways. The study utilized approaches such as network pharmacology, bioinformatics, molecular docking, and in vivo experiments. High-performance liquid chromatography (HPLC) and liquid chromatography-mass spectrometry (LC-MS) analyses revealed eight triterpenoids in G. lucidum, with ganodermanontriol being the most prominent. Molecular docking studies anticipated the interactions between these compounds and target proteins, while in vivo experiments on pneumonia-induced rat models assessed the efficacy of ganodermanontriol. Additionally, HPLC and LC-MS confirmed the presence of eight triterpenoids in the ethanol extract of G. lucidum, predominantly ganodermanontriol. Network pharmacology and molecular docking identified key genes-including TNF, EGFR, ESR1, HIF1A, HSP90AA1, and SRC-that played significant roles in the regulation of inflammatory pathways. In vivo results demonstrated that ganodermanontriol treatment mitigated lung tissue damage in rats with experimentally induced pneumonia by reducing the release of inflammatory mediators. Further mechanistic studies showed that ganodermanontriol downregulated TNF-α and inhibited the NF-κB/MAPKs signaling pathways. These findings suggested that ganodermanontriol holds promising potential as an anti-inflammatory agent for pneumonia by targeting the TNF/NF-κB/MAPKs signaling pathway, offering a novel therapeutic approach.
Collapse
Affiliation(s)
- Shizhan Deng
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Dequan Zhong
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Yonggan Dong
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Yanan Qian
- Department of Orthopedics and Traumatology, Orthopedic Trauma FacultyHenan University of Chinese MedicineZhengzhouHenanChina
| | - Biao Wang
- Wanbei Coal Electric Group General Hospital Affiliated to Bengbu Medical UniversitySuzhouAnhuiChina
| | - Mengxue Hu
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Meng Liu
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Kemeng Tan
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Chaojie Zhang
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| | - Heng Tang
- Wanbei Coal Electric Group General HospitalSuzhouAnhuiChina
| |
Collapse
|
54
|
Dong L, Lu B, Luo W, Gu X, Wu C, Trotta L, Seppanen M, Zhang Y, Zavialov AV. Intracellular concentration of ADA2 is a marker for monocyte differentiation and activation. Front Med 2025; 19:359-375. [PMID: 39832022 DOI: 10.1007/s11684-024-1110-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/18/2024] [Indexed: 01/22/2025]
Abstract
Adenosine, a critical molecule regulating cellular function both inside and outside cells, is controlled by two human adenosine deaminases: ADA1 and ADA2. While ADA1 primarily resides in the cytoplasm, ADA2 can be transported to lysosomes within cells or secreted outside the cell. Patients with ADA2 deficiency (DADA2) often suffer from systemic vasculitis due to elevated levels of TNF-α in their blood. Monocytes from DADA2 patients exhibit excessive TNF-α secretion and differentiate into pro-inflammatory M1-type macrophages. Our findings demonstrate that ADA2 localizes to endolysosomes within macrophages, and its intracellular concentration decreases in cells secreting TNF-α. This suggests that ADA2 may function as a lysosomal adenosine deaminase, regulating TNF-α expression by the cells. Interestingly, pneumonia patients exhibit higher ADA2 concentrations in their bronchoalveolar lavage (BAL), correlating with elevated pro-inflammatory cytokine levels. Conversely, cord blood has low ADA2 levels, creating a more immunosuppressive environment. Additionally, secreted ADA2 can bind to apoptotic cells, activating immune cells by reducing extracellular adenosine levels. These findings imply that ADA2 release from monocytes during inflammation, triggered by growth factors, may be crucial for cell activation. Targeting intracellular and extracellular ADA2 activities could pave the way for novel therapies in inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Liang Dong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Bingtai Lu
- Department of Respiratory Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenwen Luo
- International Center for Aging and Cancer (ICAC), Hainan Medical University, Haikou, 571199, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoqiong Gu
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
| | - Chengxiang Wu
- Tulane National Primate Research Center, Covington, USA
| | - Luca Trotta
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Mikko Seppanen
- Adult Immunodeficiency Unit, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Rare Diseases Center, Hospital for Children and Adolescents, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Yuxia Zhang
- Department of Respiratory Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, 510623, China
| | - Andrey V Zavialov
- International Center for Aging and Cancer (ICAC), Hainan Medical University, Haikou, 571199, China.
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Turku Center for Biotechnology, Turku, Finland.
| |
Collapse
|
55
|
Hiti L, Markovič T, Lainscak M, Farkaš Lainščak J, Pal E, Mlinarič-Raščan I. The immunopathogenesis of a cytokine storm: The key mechanisms underlying severe COVID-19. Cytokine Growth Factor Rev 2025; 82:1-17. [PMID: 39884914 DOI: 10.1016/j.cytogfr.2024.12.003] [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: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025]
Abstract
A cytokine storm is marked by excessive pro-inflammatory cytokine release, and has emerged as a key factor in severe COVID-19 cases - making it a critical therapeutic target. However, its pathophysiology was poorly understood, which hindered effective treatment. SARS-CoV-2 initially disrupts angiotensin signalling, promoting inflammation through ACE-2 downregulation. Some patients' immune systems then fail to shift from innate to adaptive immunity, suppressing interferon responses and leading to excessive pyroptosis and neutrophil activation. This amplifies tissue damage and inflammation, creating a pro-inflammatory loop. The result is the disruption of Th1/Th2 and Th17/Treg balances, lymphocyte exhaustion, and extensive blood clotting. Cytokine storm treatments include glucocorticoids to suppress the immune system, monoclonal antibodies to neutralize specific cytokines, and JAK inhibitors to block cytokine receptor signalling. However, the most effective treatment options for mitigating SARS-CoV-2 infection remain vaccines as a preventive measure and antiviral drugs for the early stages of infection. This article synthesizes insights into immune dysregulation in COVID-19, offering a framework to better understand cytokine storms and to improve monitoring, biomarker discovery, and treatment strategies for COVID-19 and other conditions involving cytokine storms.
Collapse
Affiliation(s)
- Luka Hiti
- Faculty of Pharmacy, University of Ljubljana, Slovenia
| | | | - Mitja Lainscak
- General Hospital Murska Sobota, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Emil Pal
- General Hospital Murska Sobota, Slovenia
| | | |
Collapse
|
56
|
Warrick KA, Vallez CN, Meibers HE, Pasare C. Bidirectional Communication Between the Innate and Adaptive Immune Systems. Annu Rev Immunol 2025; 43:489-514. [PMID: 40279312 PMCID: PMC12120936 DOI: 10.1146/annurev-immunol-083122-040624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Effective bidirectional communication between the innate and adaptive immune systems is crucial for tissue homeostasis and protective immunity against infections. The innate immune system is responsible for the early sensing of and initial response to threats, including microbial ligands, toxins, and tissue damage. Pathogen-related information, detected primarily by the innate immune system via dendritic cells, is relayed to adaptive immune cells, leading to the priming and differentiation of naive T cells into effector and memory lineages. Memory T cells that persist long after pathogen clearance are integral for durable protective immunity. In addition to rapidly responding to reinfections, memory T cells also directly instruct the interacting myeloid cells to induce innate inflammation, which resembles microbial inflammation. As such, memory T cells act as newly emerging activators of the innate immune system and function independently of direct microbial recognition. While T cell-mediated activation of the innate immune system likely evolved as a protective mechanism to combat reinfections by virulent pathogens, the detrimental outcomes of this mechanism manifest in the forms of autoimmunity and other T cell-driven pathologies. Here, we review the complexities and layers of regulation at the interface between the innate and adaptive immune systems to highlight the implications of adaptive instruction of innate immunity in health and disease.
Collapse
Affiliation(s)
- Kathrynne A Warrick
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Charles N Vallez
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Hannah E Meibers
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| |
Collapse
|
57
|
Aghaei-Zarch SM, Mahmoudieh L, Miryounesi M, Aghazadeh M, Reihani-Ardabili M, Zamani M, Motallebi M, Movafagh A. Investigation of TNF and related lncRNAs in diabetic nephropathy. Cytokine 2025; 188:156892. [PMID: 39970816 DOI: 10.1016/j.cyto.2025.156892] [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: 09/23/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a significant driver of end-stage renal disease, requiring kidney replacement therapies such as transplantation and dialysis. Given the critical importance of understanding the onset and progression of DN, we sought to explore the expression levels of tumor necrosis factor (TNF) and related long noncoding RNAs (lncRNAs) in diabetic patients with and without DN, as well as in pre-diabetic individuals, compared to healthy controls. We further explored the involvement of TNF and TNF-related lncRNAs in high glucose (HG)-induced apoptosis of human embryonic kidney (HEK)-293 cells. MATERIAL AND METHOD In the current cross-sectional investigation, we compare the expression levels of lncRNA myocardial infarction-associated transcript (MIAT), lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1), and TNF in 50 healthy individuals, 50 people with prediabetes, 50 patients with type 2 diabetes mellitus (T2DM), and 50 patients with T2DM- DN. We cultured HEK293 cells in a HG condition (100 mM glucose) to establish a cellular model of DN, while HEK293 cells cultured in a normal-glucose environment (5 mM glucose) served as controls. We further assess apoptosis in HEK293 cells via flow cytometry analysis. Moreover, we evaluated the expression levels of lncRNA MIAT, lncRNA NEAT1, and TNF in HG and normal-glucose (NG) groups to investigate their potential involvement in HEK293 cell apoptosis and the pathogenesis of DN. RESULT Our findings reveal a significant upregulation of lncRNA MIAT, lncRNA NEAT1, and TNF in T2DM and T2DM-associated DN groups compared to prediabetic individuals and healthy controls (p < 0.05). Furthermore, HG conditions significantly increased the apoptotic rate of HEK293 cells. Additionally, the expression levels of TNF, lncRNA MIAT, and lncRNA NEAT1 were increased in HEK-293 cells cultured in a HG. CONCLUSION In conclusion, our findings indicate a significant role for the TNF gene and associated lncRNAs, such as lncRNA MIAT and lncRNA NEAT1, in podocyte apoptosis and the development of DN.
Collapse
Affiliation(s)
- Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Mahmoudieh
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Miryounesi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Aghazadeh
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mehran Reihani-Ardabili
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Zamani
- Shahid Akbarabadi Clinical Research Development Unit, Iran University of medical Science, Tehran, Iran
| | - Marzieh Motallebi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
58
|
Lo CH. Targeting the inter-monomeric space of TNFR1 pre-ligand dimers: A novel binding pocket for allosteric modulators. Comput Struct Biotechnol J 2025; 27:1335-1341. [PMID: 40235642 PMCID: PMC11999085 DOI: 10.1016/j.csbj.2025.03.046] [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: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
Tumor necrosis factor (TNF) receptor 1 (TNFR1) plays a central role in signal transduction mediating inflammation and cell death associated with autoimmune and neurodegenerative disorders. Inhibition of TNFR1 signaling is a highly sought-after strategy to target these diseases. TNFR1 forms pre-ligand dimers held together by the pre-ligand assembly domain (PLAD), which is essential for receptor signaling. TNFR1 dimers form the crucial points of interaction for the entire receptor signaling complex by connecting TNF ligand bound trimeric receptors. While previous studies have shown the feasibility of disrupting TNFR1 dimeric interactions through competitive mechanism that targets the PLAD, our recent studies have demonstrated that small molecules could also bind PLAD to modulate TNFR1 signaling through an allosteric mechanism. Importantly, these allosteric modulators alter receptor dynamics and propagate long-range conformational perturbation that involves reshuffling of the receptors in the cytosolic domains without disrupting receptor-receptor or receptor-ligand interactions. In this study, we perform molecular docking of previously reported allosteric modulators on the extracellular domain of TNFR1 to understand their binding sites and interacting residues. We identify the inter-monomeric space between TNFR1 pre-ligand dimers as a novel binding pocket for allosteric modulators. We further conduct pharmacological analyses to understand the bioactivity of these compounds and their interacting residues and pharmacological properties. We then provide insights into the structure-activity relationship of these allosteric modulators and the feasibility of targeting TNFR1 conformational dynamics. This paves the way for developing new therapeutic strategies and designing chemical scaffolds to target TNFR1 signaling.
Collapse
Affiliation(s)
- Chih Hung Lo
- Department of Biology, Syracuse University, NY 13244, USA
- Interdisciplinary Neuroscience Program, Syracuse University, NY 13244, USA
| |
Collapse
|
59
|
Feng X, Xu M, Liu Y, Wang X, Duan Y, Zheng X, Yin W, Cai Y, Zhang W, Jiang Q, Pang J, Li J. The sperm quality in DIO male mice is linked to the NF-κB signaling and Ppp2ca expression in the hypothalamus. iScience 2025; 28:112110. [PMID: 40160428 PMCID: PMC11951025 DOI: 10.1016/j.isci.2025.112110] [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: 07/29/2024] [Revised: 11/24/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Recent studies show obesity correlated with reduced sperm quality in males, but the mechanism is unclear. In this study, diet-induced obese (DIO) male mice exhibited disrupted luteinizing hormone (LH) pulse release due to altered function of the hypothalamic-pituitary-gonadal (HPG) axis. This alteration was caused by activation of nuclear factor kappa B (NF-κB) signaling in the hypothalamus, which led to decreased sperm quality. RNA sequencing (RNA-seq) analysis of the hypothalamic arcuate nucleus (ARC) revealed a signaling network involving protein phosphatase 2 catalytic subunit alpha (Ppp2ca). This network disrupted LH pulse secretion by inhibiting Akt kinase (AKT) and cAMP responsive element-binding protein 1 (CREB1) activities, thereby reducing KiSS-1 metastasis-suppressor (Kiss1) expression. Furthermore, overexpression of the Ppp2ca gene in the ARC led to disrupted LH patterns and reduced sperm quality. These findings offer new insights into the molecular mechanisms underlying sperm quality decline in DIO male mice.
Collapse
Affiliation(s)
- Xu Feng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maoxing Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Liu
- Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xiaoyu Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yiman Duan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyan Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wen Yin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Wei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jing Pang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Juxue Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| |
Collapse
|
60
|
Barakat R, Al-Sarraf H, Redzic Z. Hypoxemia exerts detrimental effects on the choroid plexuses and cerebrospinal fluid system in rats. Fluids Barriers CNS 2025; 22:27. [PMID: 40075475 PMCID: PMC11905537 DOI: 10.1186/s12987-024-00613-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Hypoxemia can cause secondary acute brain injury, but the mechanisms behind it are not entirely clear and could involve disturbances in the brain extracellular fluids. We aimed to explore the effects of hypoxemia on the choroid plexus (CPs) and cerebrospinal fluid (CSF) system in rats. METHODS Male Sprague Dawley rats were kept in O2 control in vivo cabinet with either 21% (normoxia) or 8% O2 (hypoxemia) for up to 48 h. In some cases, signaling of selected cytokines was inhibited prior to hypoxemia. CSF and blood samples were collected by Cisterna Magna puncture and through venous catheters, respectively. The percentages of dead cells in the CPs and ependymal layers (EL) after hypoxemia or normoxia was estimated using TUNEL staining. CP's ultrastructure was analyzed by transmission electron microscopy. Protein concentration in the CSF and plasma was measured and the CSF albumin-to-total protein ratios were estimated. Concentrations of hypoxia-related cytokines in the CSF and plasma samples were estimated using the multiplex immunoassay. Data was analyzed by one-way ANOVA followed by either Bonferroni or Tukey's multiple comparison tests, or Student's t-test. Results are presented as mean ± SD; p < 0.05 was considered statistically significant. RESULTS Duration of hypoxemia exerted significant effects on the cell viability in the CPs (p < 0.01) and EL (p < 0.01) and caused apoptosis-related changes in the CP. Hypoxemia had significant effects on the protein concentration in the CSF (p < 0.05), but not in plasma (p > 0.05), with a significant increase in the CSF albumin-to-total protein ratio after 6 h hypoxemia (p < 0.05). Thirty-two cytokines were detected in the CSF. Hypoxemia caused a statistically significant reduction in the concentrations of 12 cytokines, while concentrations of erythropoietin (EPO) and vascular endothelial growth factor (VEGF) increased significantly. Exposure to hypoxemia after inhibitions of EPO, VEGF, or tumor necrosis factor alpha (TNFα) signaling resulted in more dead cells (p < 0.01), less dead cells (p < 0.01) and more dead cells (p < 0.01) in the CPs, respectively, when compared to the number of dead cells when these cytokines were not inhibited. The density of macrophages in the CPs decreased significantly during hypoxemia; that effect was cancelled out by TNFα inhibition. CONCLUSION Hypoxemia had detrimental effects on the CPs and CSF system, which was modulated by hypoxia- and inflammation-related cytokines.
Collapse
Affiliation(s)
- Rawan Barakat
- Department of Physiology, College of Medicine, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait
| | - Hameed Al-Sarraf
- Department of Physiology, College of Medicine, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait
| | - Zoran Redzic
- Department of Physiology, College of Medicine, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait.
| |
Collapse
|
61
|
Saha S, Roy S, Hazra A, Das D, Kumar V, Singh AK, Singh AV, Mondal R, Bose Dasgupta S. S-nitrosylation-triggered secretion of mycobacterial PknG leads to phosphorylation of SODD to prevent apoptosis of infected macrophages. Proc Natl Acad Sci U S A 2025; 122:e2404106122. [PMID: 40035756 PMCID: PMC11912491 DOI: 10.1073/pnas.2404106122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
The tuberculosis-causing agent Mycobacterium tuberculosis (M.tb) establishes its niche inside macrophages by secretion of several virulence factors and engaging many host factors. Mycobacterial infection of macrophages results in a proinflammatory trigger-mediated secretion of TNFα. Protein kinase G (PknG), a Serine/Threonine kinase, is essential for mycobacterial survival within the macrophage. Pathogenic mycobacteria, upon infection, can trigger the secretion of proinflammatory cytokine TNFα, but whether secreted PknG plays any role in TNFα secretion at early stages of infection remains undeciphered. Moreover, at early infection stages, prevention of macrophage apoptosis is vital to successful mycobacterial pathogenesis. Our studies show that mycobacteria-secreted PknG can dampen the expression and concomitant secretion of proinflammatory TNFα. During early infection, M.tb infection-induced generation of reactive nitrogen intermediates (RNI) leads to S-nitrosylation of PknG on Cys109, thereby enabling its secretion into macrophages. Upon M.tb infection, secreted S-nitrosylated PknG phosphorylates macrophage Silencer of Death Domains (SODD) at Thr405, as identified through our phosphoproteomic studies. Thereafter, phosphorylated SODD, through an irreversible binding with the TNFR1 death domain, prevents Caspase8 activation and concomitant extrinsic apoptotic trigger. Moreover, alveolar macrophages from mice infected with PknG-knockout M.tb also exhibited SODD phosphorylation and hindered Caspase8 activation to prevent extrinsic macrophage apoptosis. Therefore, this work exhibits S-nitrosylation-mediated secretion of PknG to induce phosphorylation of macrophage SODD, which, through irreversible interaction with TNFR1, prevented extrinsic macrophage apoptosis at the early stages of infection.
Collapse
Affiliation(s)
- Saradindu Saha
- Molecular Immunology and Cellular Microbiology Laboratory, Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur721302, India
| | - Sadhana Roy
- Molecular Immunology and Cellular Microbiology Laboratory, Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur721302, India
| | - Arnab Hazra
- Molecular Immunology and Cellular Microbiology Laboratory, Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur721302, India
| | - Debayan Das
- Molecular Immunology and Cellular Microbiology Laboratory, Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur721302, India
| | - Vimal Kumar
- Laboratory for Animal Experiments, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra282006, India
| | - Amit Kumar Singh
- Laboratory for Animal Experiments, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra282006, India
| | - Ajay Vir Singh
- Department of Microbiology and Molecular Biology, Indian Council of Medical Research (ICMR)-National Japanese Leprosy Mission for Asia (JALMA) Institute for Leprosy and Other Mycobacterial Diseases, Agra282006, India
| | - Rajesh Mondal
- Department of Bacteriology, National Institute for Research in Tuberculosis, Chennai, Tamil Nadu600031, India
- ICMR—National Institute for Research in Environmental Health, Bhopal462030, India
| | - Somdeb Bose Dasgupta
- Molecular Immunology and Cellular Microbiology Laboratory, Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur721302, India
| |
Collapse
|
62
|
Sun Y, Liu L, He H, Cui G, Zheng Y, Ye C, Qu L, Sun Y, Ji J, Lammers T, Zhang Y, Zhong Z. Co-activating STING-TLR9 pathways promotes radiotherapy-induced cancer vaccination. J Control Release 2025; 379:327-343. [PMID: 39778743 DOI: 10.1016/j.jconrel.2024.12.079] [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: 11/08/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Vaccination may cure cancer patients by inducing tumor-specific immune responses. Radiotherapy is an appealing strategy to generate cancer vaccines in situ; thus far, however, only modest and short-lived immune responses are achieved. We here show that radiation combined with co-activating STING-TLR9 can generate powerful in situ cancer vaccines. Notably, radiation at a dose of 12Gy is found to be optimal for boosting tumor cell immunogenicity, and STING-TLR9 co-stimulation by a dual immune activation nano-agonist overrides key immunosuppressive effects associated with radiotherapy. Local radiotherapy combined with the dual immune activation nano-agonists elicits strong systemic anti-tumor immune responses, resulting in complete regression of tumors and metastases in multiple syngeneic murine tumor models. This work introduces a novel and highly potent cancer immunotherapeutic strategy that holds promise for the personalized treatment of intractable cancers.
Collapse
Affiliation(s)
- Yu Sun
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Liang Liu
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; School of Optical and Electronic Information, Suzhou City University, Suzhou 215104, People's Republic of China
| | - Huilan He
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanhong Cui
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Yun Zheng
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Chunlian Ye
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Liping Qu
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Yinping Sun
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Jinlong Ji
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Clinic, Aachen 52074, Germany
| | - Ying Zhang
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
63
|
Mahjoubin-Tehran M, Rezaei S, Butler AE, Sahebkar A. Decoy oligonucleotides targeting NF-κB: a promising therapeutic approach for inflammatory diseases. Inflamm Res 2025; 74:47. [PMID: 40047902 DOI: 10.1007/s00011-025-02021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 05/13/2025] Open
Abstract
Nuclear factor-kappa B (NF-κB) transcription factor plays a crucial function in controlling several cellular processes, including the production of inflammatory mediators. The aberrant activation of this transcription factor and its signaling pathway is associated with the pathophysiology of many diseases. Therefore, discovering drugs that target NF-κB is crucial for treating various diseases. Decoy oligonucleotides (decoy ONs) are a pharmacological approach that specifically inhibits NF-κB activation and are used to treat several inflammatory diseases. Decoys that target NF-κB have been shown to enhance radiosensitivity and drug sensitivity in vitro and strongly block IL-6 and IL-8 gene expression induced by TNF-α in experimental cell systems. In vivo, NF-κB decoy reduced atherosclerotic plaque, prevented atopic dermatitis and extended cardiac transplant survival. Decoys have the potential to be used in clinical applications, but they face several challenges. To overcome these limitations, researchers have conducted studies on chemical modifications and delivery techniques. Innovative compounds that target NF-κB, such as NF-κB-decoy-based sensor-containing models, phosphorothioate hairpin-modified oligonucleotides, and peptide nucleic acid (PNA)-based transcription factor decoys, are very attractive. This research aims to explore the use of decoys to combat NF-κB in various disorders.
Collapse
Affiliation(s)
| | - Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - 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
|
64
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
65
|
Lai GY, Lee YC, Weng HJ, Lai KH, Hsiang MC, Hsu KY, Liao CP. Discoidin domain receptor inhibitor DDR1-IN-1 induces autophagy and necroptotic cell death in malignant peripheral nerve sheath tumor. Cell Death Discov 2025; 11:83. [PMID: 40025071 PMCID: PMC11873111 DOI: 10.1038/s41420-025-02367-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a soft tissue sarcoma commonly associated with the tumor-predisposition disorder neurofibromatosis 1. The extracellular matrix collagens contribute to many fibrotic tumors; however, the role of collagen signaling in MPNST was unclear. This study investigated the effects of blocking the interaction between collagens and their receptors in MPNST. We first analyzed the expressions of collagen family proteins in MPNSTs and found an overall increase compared to neurofibroma. Treatment of DDR1-IN-1, a small molecule inhibitor for the collagen receptor discoidin domain receptor, induced a robust MPNST cell death, highlighting the dependence of MPNST survival on collagen signaling. DDR1-IN-1 induced MPNST cell death by activating autophagy and necroptosis signaling. Treatment of necroptosis inhibitors necrostatin-1 or necrosulfonamide reduced the numbers of DDR1-IN-1-induced necrotic cells and autolysosomes, suggesting that the autophagic process depends on necroptosis activation. Combinations of DDR1-IN-1 with other anti-MPNST agents revealed synergistic activities against MPNST. In summary, this study discovered a critical MPNST death signaling induced by the small molecule DDR1-IN-1, which might shed light on future MPNST therapeutic strategies.
Collapse
Affiliation(s)
- Guan-Yi Lai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Cheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hao-Jui Weng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Dermatology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, 23561, Taiwan
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Min-Chen Hsiang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kai-Yu Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Ping Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Cancer Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
| |
Collapse
|
66
|
Millar CL, Iloputaife I, Baldyga K, Norling AM, Boulougoura A, Vichos T, Tchkonia T, Deisinger A, Pirtskhalava T, Kirkland JL, Travison TG, Lipsitz LA. A pilot study of senolytics to improve cognition and mobility in older adults at risk for Alzheimer's disease. EBioMedicine 2025; 113:105612. [PMID: 40010154 PMCID: PMC11907475 DOI: 10.1016/j.ebiom.2025.105612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND This single-arm study evaluates the feasibility, safety, and preliminary effects of two senolytic agents, Dasatinib and Quercetin (DQ), in older adults at risk of Alzheimer's disease. METHODS Participants took 100 mg of Dasatinib and 1250 mg of Quercetin for two days every two weeks over 12 weeks. Recruitment rate, adverse events, absolute changes in functional outcomes, and percent changes in biomarkers were calculated. Spearman correlations between functional and biomarker outcomes were performed. FINDINGS Approximately 10% of telephone-screened individuals completed the intervention (n = 12). There were no serious adverse events related to the intervention. Mean Montreal Cognitive Assessment (MoCA) scores non-significantly increased following DQ by 1.0 point (95% CI: -0.7, 2.7), but increased significantly by 2.0 points (95% CI: 0.1, 4.0) in those with lowest baseline MoCA scores. Mean percent change in tumour necrosis factor-alpha (TNF-α), a key product of the senescence-associated secretory phenotype (SASP), non-significantly decreased following DQ by -3.0% (95% CI: -13.0, 7.1). Changes in TNF-α were significantly and inversely correlated with changes in MoCA scores (r = -0.65, p = 0.02), such that reductions in TNF- α were correlated with increases in MoCA scores. INTERPRETATION This study suggests that intermittent DQ treatment is feasible and safe; data hint at potential functional benefits in older adults at risk of Alzheimer's disease. The observed reduction in TNF-α and its correlation with increases in MoCA scores suggests that DQ may improve cognition by modulating the SASP. However, there was not an appropriate control group. Data are preliminary and must be interpreted cautiously. FUNDING National Institute on Ageing grants R21AG073886 and R33AG061456 funded this research.
Collapse
Affiliation(s)
- Courtney L Millar
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, 1200 Centre St, Boston, MA 02131, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA.
| | - Ike Iloputaife
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, 1200 Centre St, Boston, MA 02131, USA
| | - Kathryn Baldyga
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, 1200 Centre St, Boston, MA 02131, USA
| | - Amani M Norling
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, 1200 Centre St, Boston, MA 02131, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA
| | - Afroditi Boulougoura
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Division of Rheumatology & Clinical Immunology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA
| | - Theodoros Vichos
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Division of Rheumatology & Clinical Immunology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA
| | - Tamara Tchkonia
- Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Aaron Deisinger
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Tamar Pirtskhalava
- Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - James L Kirkland
- Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Thomas G Travison
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, 1200 Centre St, Boston, MA 02131, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA
| | - Lewis A Lipsitz
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, 1200 Centre St, Boston, MA 02131, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA
| |
Collapse
|
67
|
Liu Y, Chen X, Chen J, Song C, Wei Z, Liu Z, Liu F. The Significance of MAPK Signaling Pathway in the Diagnosis and Subtype Classification of Intervertebral Disc Degeneration. JOR Spine 2025; 8:e70060. [PMID: 40134951 PMCID: PMC11932887 DOI: 10.1002/jsp2.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IDD) is a human aging disease related mainly to inflammation, cellular senescence, RNA/DNA methylation, and ECM. The mitogen-activated protein kinase (MAPK) signaling pathway is engaged in multiple biological functions by phosphorylating specific serine and threonine residues on target proteins through phosphorylation cascade effects, but the role and specific mechanisms of the MAPK signaling pathway in IDD are still unclear. METHODS We identified 20 MAPK-related differential genes by differential analysis of the GSE124272 and GSE150408 datasets from the GEO database. To explore the biological functions of these differential genes in humans, we performed GO and KEGG analyses. Additionally, we applied PPI networks, LASSO analysis, the RF algorithm, and the SVM-RFE algorithm to identify core MAPK-related genes. Finally, we conducted further validation using clinical samples. RESULTS We ultimately identified and validated four pivotal MAPK-related genes, namely, KRAS, JUN, RAP1B, and TNF, using clinical samples, and constructed the ROC curves to evaluate the predictive accuracy of the hub genes. A nomogram model was subsequently developed based on these four hub MAPK genes to predict the prevalence of IDD. Based on these four hub genes, we classified IDD patients into two MAP clusters by applying the consensus clustering method and identified 1916 DEGs by analyzing the differences between the two clusters. Further analysis using the same approach allowed us to identify two gene clusters based on these DEGs. We used a PCA algorithm to determine the MAPK score for each sample and discovered that MAPK cluster A and gene cluster A had higher scores, suggesting greater sensitivity to MAPK signaling pathway-associated agents in the subtype. We displayed the differing expression levels of four hub MAPK-related genes across the two clusters and their relationship with immune cell infiltration to highlight the distinctions between clusters A and B. CONCLUSION In summary, four hub MAPK signaling pathway-related genes, KRAS, JUN, RAP1B, and TNF, could be applied to the diagnosis and subtype classification of IDD and benefit the prevention and treatment of IDD.
Collapse
Affiliation(s)
- Yong Liu
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese MedicineSouthwest Medical UniversityLuzhouChina
| | - Xueyan Chen
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Jingwen Chen
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese MedicineSouthwest Medical UniversityLuzhouChina
| | - Chao Song
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese MedicineSouthwest Medical UniversityLuzhouChina
| | - Zhangchao Wei
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese MedicineSouthwest Medical UniversityLuzhouChina
| | - Zongchao Liu
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese MedicineSouthwest Medical UniversityLuzhouChina
- Department of OrthopedicsLuzhou Longmatan District People's HospitalLuzhouChina
| | - Fei Liu
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese MedicineSouthwest Medical UniversityLuzhouChina
- Department of OrthopedicsRuiKang Hospital Affiliated to Guangxi University of Chinese MedicineNanningChina
| |
Collapse
|
68
|
Christiansen JR, Ferreira SA, Szymkowski DE, Jakobsson J, Tansey MG, Romero-Ramos M. Peripherally administered TNF inhibitor is not protective against α-synuclein-induced dopaminergic neuronal death in rats. Neurobiol Dis 2025; 206:106803. [PMID: 39800228 DOI: 10.1016/j.nbd.2025.106803] [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: 10/07/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
The underlying cause of neuronal loss in Parkinson's disease (PD) remains unknown, but evidence implicates neuroinflammation in PD pathobiology. The pro-inflammatory cytokine soluble tumor necrosis factor (TNF) seems to play an important role and thus has been proposed as a therapeutic target for modulation of the neuroinflammatory processes in PD. In this regard, dominant-negative TNF (DN-TNF) agents are promising antagonists that selectively inhibit soluble TNF signaling, while preserving the beneficial effects of transmembrane TNF. Previous studies have tested the protective potential of DN-TNF-based therapy in toxin-based PD models. Here we test for the first time the protective potential of a DN-TNF therapeutic against α-synuclein-driven neurodegeneration in the viral vector-based PD female rat model. To do so, we administered the DN-TNF agent XPro1595 subcutaneously for a period of 12 weeks. In contrast to previous studies using different PD models, neuroprotection was not achieved by systemic XPro1595 treatment. α-Synuclein-induced loss of nigrostriatal neurons, accumulation of pathological inclusions and microgliosis was detected in both XPro1595- and saline-treated animals. XPro1595 treatment increased the percentage of the hypertrophic/ameboid Iba1+ cells in SN and reduced the striatal MHCII+ expression in the α-synuclein-overexpressing animals. However, the treatment did not prevent the MHCII upregulation seen in the SN of the model, nor the increase of CD68+ phagocytic cells. Therefore, despite an apparently immunomodulatory effect, this did not suffice to protect against viral vector-derived α-synuclein-induced neurotoxicity. Further studies are warranted to better elucidate the therapeutic potential of soluble TNF inhibitors in PD.
Collapse
Affiliation(s)
- Josefine R Christiansen
- Department of Biomedicine & Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, 8000 Aarhus, Denmark
| | - Sara A Ferreira
- Department of Biomedicine & Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, 8000 Aarhus, Denmark
| | | | - Johan Jakobsson
- Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, Lund University, BMC A11 and B10, S-221 84 Lund, Sweden
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, United States of America
| | - Marina Romero-Ramos
- Department of Biomedicine & Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, 8000 Aarhus, Denmark.
| |
Collapse
|
69
|
Lee W, An G, Kim J, Lee H, Song G, Lim W, Jeong W. Evaluation of thiobencarb herbicide-induced cytotoxicity mediated via disruption of calcium homeostasis in bovine mammary glands: A comprehensive in vitro and in silico study. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 208:106267. [PMID: 40015859 DOI: 10.1016/j.pestbp.2024.106267] [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: 09/23/2024] [Revised: 11/17/2024] [Accepted: 12/17/2024] [Indexed: 03/01/2025]
Abstract
In contemporary agriculture, the predominant strategy for managing perennial weeds within agroecosystems involves the extensive use of herbicides. Thiobencarb is widely employed to control gramineous weeds during rice cultivation. This herbicide is commonly found in terrestrial environments and agricultural products. The harmful potential of thiobencarb has been reported, along with its adverse effects in exposed species. However, few studies have explored thiobencarb toxicity, specifically in dairy cows, despite the possibility of ingestion through soil residues. Exposure to xenobiotics can reduce the viability or impair the function of bovine mammary epithelial cells (BMECs), leading to compromised immune function and reduced milk production. Despite the known cytotoxicity of thiobencarb, its specific effects on BMECs remain unclear. Herein, we aimed to investigate the effect of thiobencarb on milk production by examining its toxic effects and underlying mechanisms in BMECs. We assessed the cytotoxic effects of thiobencarb and analyzed various cellular responses upon exposure. Thiobencarb-induced apoptosis was associated with disrupted calcium homeostasis. Additionally, thiobencarb modulated AKT/MAPK proteins and increased mRNA levels of genes related to the inflammatory response. Furthermore, treatment of BMECs with thiobencarb suppressed the expression of genes related to milk production, including those encoding superoxide dismutase, tight junctions, and casein. Finally, we conducted an in silico molecular docking analysis to evaluate the binding affinity between thiobencarb and target proteins.
Collapse
Affiliation(s)
- Woonghee Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, San Diego, CA, United States; Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jinyoung Kim
- Department of Animal Biotechnology, Dankook University, Cheonan 31116, Republic of Korea
| | - Hojun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Wooyoung Jeong
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung 25601, Republic of Korea.
| |
Collapse
|
70
|
Miao X, Zhou X, Liu C, Shi H, Liu F, Ma Y, Shi H. Alcohol-Induced Dendritic Cells and Their Exosomes Promote T-Cell Immunity in Hepatitis B Virus Transgenic Mice and Patients With Chronic Hepatitis B. J Med Virol 2025; 97:e70287. [PMID: 40045507 DOI: 10.1002/jmv.70287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/20/2025] [Accepted: 02/19/2025] [Indexed: 05/13/2025]
Abstract
Dendritic cells and the exosomes they secrete play a crucial role in the immune system, and studies have shown that dendritic cell function is dramatically reduced in patients with chronic hepatitis B. Alcohol could stimulate dendritic cell maturation. Consequently, the present work explored the therapeutic effect of alcohol-induced dendritic cells and their exosomes in hepatitis B virus (HBV) infection. We systematically investigated the functional effects of alcohol stimulation and HBV infection on dendritic cells and their exosomes, as well as cocultured alcohol-induced dendritic cells and exosomes with lymphocytes from HBV transgenic mice and chronic hepatitis B patients to study the T cell immune response. Our findings revealed that alcohol significantly accelerated the maturation of bone marrow-derived dendritic cells in mice and dendritic cells in patients with chronic hepatitis B, but had no effect on the DC2.4 cell line. Simultaneously, HBV infection was demonstrated to inhibit dendritic cell activation and maturation, as well as exosomes. More importantly, alcohol-induced dendritic cells enhanced T-cell immunity in HBV transgenic mice and chronic hepatitis B patients, and their exosomes had the same impact. The maturation of dendritic cells and their exosomes can be effectively induced by alcohol. Meanwhile, alcohol-induced maturation of dendritic cells and exosomes can significantly repair the poor T-cell immunity caused by HBV infection, making it a promising novel treatment for chronic hepatitis B patients in the future.
Collapse
Affiliation(s)
- Xingzhong Miao
- Beijing Key Laboratory Research on Liver Regeneration and Artificial Liver Transformation, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi, China
| | - Chaonan Liu
- Beijing Key Laboratory Research on Liver Regeneration and Artificial Liver Transformation, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Department of Nephrology, Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi, China
| | - Honglin Shi
- Beijing Key Laboratory Research on Liver Regeneration and Artificial Liver Transformation, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Fang Liu
- Beijing Key Laboratory Research on Liver Regeneration and Artificial Liver Transformation, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Beijing Key Laboratory Research on Liver Regeneration and Artificial Liver Transformation, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hongbo Shi
- Beijing Key Laboratory Research on Liver Regeneration and Artificial Liver Transformation, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
71
|
Weng J, Geng M, Hu X, Hu Y, Yang Y, Xing B, Wu Z, Wei Z. Design of minibinder proteins specific to TNFR1. Int J Biol Macromol 2025; 293:139403. [PMID: 39743082 DOI: 10.1016/j.ijbiomac.2024.139403] [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: 11/11/2024] [Revised: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
TNFα inhibitors have been successfully developed and used in the clinical treatment of autoimmune disorders. However, the use of TNFα blockade may be accompanied by side effects. The cases of bacterial and viral infections, lymphoproliferative disorders, and anti-TNFα-induced lupus, have been reported among the rheumatoid arthritis or Crohn's disease patients treated with TNFα blockers. Therefore, alternative therapeutic strategy is highly desirable. TNFα signaling via TNFR1 induces proinflammatory responses, and selective inhibition of TNFR1 signaling may be beneficial for managing autoimmune diseases. To this end, we developed minibinder proteins targeting soluble ectodomain of TNFR1 (sTNFR1) by de novo computational designing. Top-rated designed minibinders targeting to two hydrophobic sites on sTNFR1 were selected and expressed in E. coli. Purified top-ranked minibinders are thermostable in solution and presented micromolar to sub-micromolar affinity to sTNFR1. All designs showed the potency of blocking TNFα signaling in L929 cell-based assays, and one of the designs targeting to hydrophobic Site I called S1B2 exhibited higher potency than other minibinders with IC50 of 4.32 nM. Our work provided a new approach to develop TNFR1 antagonist and S1B2 should be a promising lead molecule of TNFR1 antagonist for further development.
Collapse
Affiliation(s)
- Jun Weng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Miaomiao Geng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Xiaoyu Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yang Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yi Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Banbin Xing
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Zhiyong Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Zigong Wei
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China; Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China.
| |
Collapse
|
72
|
Wang J, Shi H, Yang Y, Gong X. Crosstalk between ferroptosis and innate immune in diabetic kidney disease: mechanisms and therapeutic implications. Front Immunol 2025; 16:1505794. [PMID: 40092979 PMCID: PMC11906378 DOI: 10.3389/fimmu.2025.1505794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent complication of diabetes mellitus (DM), and its incidence is increasing alongside the number of diabetes cases. Effective treatment and long-term management of DKD present significant challenges; thus, a deeper understanding of its pathogenesis is essential to address this issue. Chronic inflammation and abnormal cell death in the kidney closely associate with DKD development. Recently, there has been considerable attention focused on immune cell infiltration into renal tissues and its inflammatory response's role in disease progression. Concurrently, ferroptosis-a novel form of cell death-has emerged as a critical factor in DKD pathogenesis, leading to increased glomerular filtration permeability, proteinuria, tubular injury, interstitial fibrosis, and other pathological processes. The cardiorenal benefits of SGLT2 inhibitors (SGLT2-i) in DKD patients have been demonstrated through numerous large clinical trials. Moreover, further exploratory experiments indicate these drugs may ameliorate serum and urinary markers of inflammation, such as TNF-α, and inhibit ferroptosis in DKD models. Consequently, investigating the interplay between ferroptosis and innate immune and inflammatory responses in DKD is essential for guiding future drug development. This review presents an overview of ferroptosis within the context of DKD, beginning with its core mechanisms and delving into its potential roles in DKD progression. We will also analyze how aberrant innate immune cells, molecules, and signaling pathways contribute to disease progression. Finally, we discuss the interactions between ferroptosis and immune responses, as well as targeted therapeutic agents, based on current evidence. By analyzing the interplay between ferroptosis and innate immunity alongside its inflammatory responses in DKD, we aim to provide insights for clinical management and drug development in this area.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Geriatric Integrative, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Haonan Shi
- School of Medicine, Shanghai University, Shanghai, China
| | - Ye Yang
- Department of Geriatric Integrative, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xueli Gong
- Department of Pathophysiology, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
73
|
Yamamoto T, Mitsunaga F, Kotani A, Tajima K, Wasaki K, Nakamura S. Safety Assessment of a Sublingual Vaccine Formulated with Poly(I:C) Adjuvant and Influenza HA Antigen in Mice and Macaque Monkeys: Comparison with Intranasal Vaccine. Vaccines (Basel) 2025; 13:261. [PMID: 40266106 PMCID: PMC11945353 DOI: 10.3390/vaccines13030261] [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: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
A sublingual vaccine comprising the Poly(I:C) adjuvant and influenza HA antigen was evaluated for safety in both mice and macaque monkeys relative to its intranasal counterpart. Safety was assessed in terms of harmful effects corresponding to the upregulation of the inflammation-associated genes Saa3, Tnf, IL6, IL1b, Ccl2, Timp1, C2, Ifi47, Aif1, Omp, Nos2, and/or Gzmb in mice and SAA2, TNF, IL6, IL1B, CCL2, TIMP, C2, AIF1, and GZMB in macaques. Quantitative gene expression analyses were performed using RT-qPCR with RNA samples from four tissue types, the olfactory bulb, pons, lung, tongue, and lymph node, from both mice and macaques. In mice, the intranasally delivered vaccine markedly upregulated the inflammation-related genes in the olfactory bulb 1 day and 7 days after vaccination. The adverse effects of intranasal vaccination were also observed in macaques, albeit to a lesser extent than in mice. The intranasal vaccination also upregulated these genes in the pons of both mice and macaques. In contrast, the sublingual vaccine did not adversely affect the olfactory bulb or pons in either mice or macaques. The intranasally administered vaccine significantly upregulated these genes in the lungs only 1 day after vaccination, but not 7 days later, in both mice and macaques. We conclude that intranasal vaccination results in unfavorable side effects corresponding to upregulated inflammatory genes in the brain (olfactory bulb and pons). Sublingual vaccination, however, did not induce these side effects in either mice or macaques and was hence evaluated as safe.
Collapse
Affiliation(s)
- Tetsuro Yamamoto
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- EP Mediate Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
- Research Center, EPS Innovative Medicine Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Fusako Mitsunaga
- Intelligence & Technology Lab, Inc., 52-1 Fukue, Kaizu-cho, Kaizu 503-0628, Japan;
- Biomedical Institute, NPO Primate Agora, 52-2 Fukue, Kaizu-cho, Kaizu 503-0628, Japan
| | - Atsushi Kotani
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- Research Center, EPS Innovative Medicine Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Kazuki Tajima
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- Research Center, EPS Innovative Medicine Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Kunihiko Wasaki
- Innovation Research Center, EPS Holdings, Inc., 2-1 Tsukudohachimancho, Shinjuku-ku, Tokyo 162-0815, Japan; (T.Y.); (A.K.); (K.T.); (K.W.)
- EP Mediate Co., Ltd., 1-8 Tsukudocho, Shinjuku-ku, Tokyo 162-0821, Japan
| | - Shin Nakamura
- Intelligence & Technology Lab, Inc., 52-1 Fukue, Kaizu-cho, Kaizu 503-0628, Japan;
- Biomedical Institute, NPO Primate Agora, 52-2 Fukue, Kaizu-cho, Kaizu 503-0628, Japan
| |
Collapse
|
74
|
Wang L, Dong Y, Yang Q, Liu S, Wu B, Zhang D, Shen S, Xin C, Liu Z, Wu Q, Huang G, Duan L. Exploring the association between rheumatoid arthritis and non-small cell lung cancer risk: a transcriptomic and drug target-based analysis. Hereditas 2025; 162:28. [PMID: 40016789 PMCID: PMC11866852 DOI: 10.1186/s41065-025-00396-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a common subtype of lung cancer that has received considerable attention for its potential association with rheumatoid arthritis (RA). However, current understanding of the relationship between RA and NSCLC risk remains limited and in-depth studies of molecular mechanisms are lacking. METHODS We obtained transcriptomic data of NSCLC from the Gene Expression Omnibus (GEO) database and performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of differential genes. We then used Mendelian randomisation (MR) analysis to explore the causal relationship between RA and NSCLC, but the results showed no direct causal relationship between RA and NSCLC. In light of this finding, we shifted our research focus to investigate the effect of RA therapeutics on NSCLC risk. A drug-targeted MR analysis of drugs available for the treatment of RA was performed by searching for drugs that target NSCLC differential genes associated with RA. RESULTS We found that several of the drugs corresponding to NSCLC differential genes associated with RA are used to treat RA. By drug-targeted MR analysis of drugs, we found that some drugs do have an effect on the risk of developing NSCLC, increasing the risk of developing NSCLC. CONCLUSION This study employed transcriptomic analysis and MR of drug targets to elucidate the potential correlation between RA and the risk of developing NSCLC. The identification of NSCLC differentially expressed genes associated with RA and their drug targets has provided new perspectives for an in-depth understanding of the pathogenesis of NSCLC. Furthermore, an additional immune infiltration analysis demonstrated that, in NSCLC tissues, the infiltration levels of specific immune cell subpopulations, including regulatory T cells (Tregs), activated natural killer cells (NK cells) and unpolarised macrophages (M0), exhibited notable differences. These findings emphasise the significant role that immune cell interactions between RA and NSCLC may play in disease progression. Furthermore, through the analysis of validation histology, we have further confirmed the potential role of differential genes associated with RA in the development of NSCLC. The expression levels of these genes demonstrated significant differences in NSCLC samples, providing a basis for possible future therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Lyubo Wang
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Yuxian Dong
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qingcheng Yang
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Siyun Liu
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Bencheng Wu
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Dahang Zhang
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Shuai Shen
- First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Chenjun Xin
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Zurui Liu
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Qiuyang Wu
- Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming(in Yunnan), China
| | - Guojian Huang
- Kunming Dongchuan District People's Hospital, Kunming, Yunnan, China
| | - Lincan Duan
- Pu'er People's Hospital, Pu'er, Yunnan, China.
| |
Collapse
|
75
|
Pareek D, Zeyaullah M, Patra S, Alagu O, Singh G, Wasnik K, Gupta PS, Paik P. Mesoporous polymeric nanoparticles for effective treatment of inflammatory diseases: an in vivo study. J Mater Chem B 2025; 13:3094-3113. [PMID: 39902477 DOI: 10.1039/d4tb02012j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Acute inflammatory diseases require suitable medicine over the existing therapeutics. In this line, the present work is focused on developing polymeric nanomedicine for the treatment of inflammatory disorders. Herein, cell viable nanoparticles (GlyNPs) of size 180-250 nm in diameter and pore size of 4-5 nm in diameter, based on glycine and acryloyl chloride, have been developed and proved to be a potential anti-inflammatory agent without using any conventional drugs. These particles exhibit colloidal stability (with a zeta potential of -35.6 mV). A network pharmacology-based computational study has been executed on 9076 genes and proteins responsible for inflammatory diseases, out of which 10 are selected that have a major role in rheumatoid arthritis (RA). In silico docking study has been conducted to find out the targeted efficiency of the GlyNPs considering 10 inflammation-specific markers, namely IL-6, IL-1β, TNF-α, TLR-4, STAT-1, MAPK-8, MAPK-14, iNOS, NF-κβ and COX-2. The results revealed that the GlyNPs could be an excellent anti-inflammatory component similar to aspirin. The in vitro inflammation activity of these GlyNPs has also been checked on an inflammation model generated by LPS in RAW 264.7 macrophages. Then, the in vitro anti-inflammation efficiency has been checked with 10-150 μg mL-1 of GlyNP doses. The treatment efficiency has been checked on inflammation-responsible immune markers (NO level, NF-κβ, INF-γ, IL-6, IL-10, and TNF-α) and it was found that the GlyNPs are an excellent component in reducing inflammation. The in vivo therapeutic response of GlyNPs on the induced rheumatoid arthritis (RA) model has been evaluated by measuring the morphological, biochemical and immune-cytokine and interferon levels responsible for the inflammation, using a 2 g kg-1 dose (sample to weight of rat). The anti-inflammatory efficiency of GlyNPs without using additional drugs was found to be excellent. Thus, GlyNPs could be paramount for the potential treatment of various inflammatory diseases.
Collapse
Affiliation(s)
- Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Md Zeyaullah
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Oviya Alagu
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Gurmeet Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| |
Collapse
|
76
|
Yang X, Fang R, Li X, Kong W, Jin Y, Jiao R, Liu Z, Zhang M, Peng Q, Zhang Y, Song N. Engineered Nanovesicles for the Precise and Noninvasive Treatment of Deep Osteomyelitis Caused by MRSA Infection with Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11795-11810. [PMID: 39945439 DOI: 10.1021/acsami.4c20893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The clinical treatment of hospital-acquired persistent osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) presents two major challenges: ineffective drug delivery into deep tissues and counteracting the rapid establishment of an immunosuppressive microenvironment. Indeed, MRSA can evade immunosurveillance and undermine both innate and adaptive immune responses. Herein, the engineered nanovesicles, functioning by combining sonodynamic therapy (SDT) with immune modulation, were constructed for the precise and noninvasive removal of MRSA in deep tissue and activation of the antimicrobial immune response using a newly engineered nanovesicle. Macrophage-derived M1 phenotypic microvesicles (M1-MW) internalized vancomycin-cross-linked micelles with the acoustic sensitizer indocyanine green (ICG) (VCG micelles). The vesicles of M1-MW were grafted with PEGylated mannose, allowing for targeted accumulation at the infection site. The VCG micelles were responsive to the highly reducing environment and released ICG to generate ROS after exposure to ultrasounds. This effect was combined with the presence of vancomycin to kill MRSA. In an osteomyelitis infection model, we observed an improved survival rate and reprogramming of macrophages to a pro-inflammatory M1 phenotype. The latter promoted T-cell activation and immune defense against MRSA-camouflaged homologous cell-transferred infections. Thus, our study presents a noninvasive and efficient treatment (VCG@MMW) for deep osteomyelitis with improved bacterial clearance and reduced risk of recurrence with enhanced immune response.
Collapse
Affiliation(s)
- Xingyue Yang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Weihao Kong
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yubao Jin
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ruohan Jiao
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Zhenggong Liu
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Meiqi Zhang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Qixian Peng
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, P. R. China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| |
Collapse
|
77
|
Koru I, Atasever-Arslan B. Protective effects of sesamol against cigarette smoke toxicity on the blood-brain barrier. BMC Complement Med Ther 2025; 25:68. [PMID: 39987119 PMCID: PMC11846467 DOI: 10.1186/s12906-025-04796-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/29/2025] [Indexed: 02/24/2025] Open
Abstract
Cigarette smoke comprises nicotine, reactive oxygen species (ROS), and carcinogens, which can induce oxidative stress and inflammation, leading to disruption of the blood-brain barrier. This study utilized cigarette smoke extract (CSE) in an in vitro model of the blood-brain barrier (BBB).Sesamol is a phenolic compound derived from Sesamum indicum L. Its potential to reduce inflammation and provide protection was also examined. As a result of the study, it was found that CSE significantly increases permeability by degrading the BBB, whereas a protective effect was observed in the sesamol-incubated group within the BBB model. While the Sesamol + CSE group does not entirely prevent the damage induced by CSE in the barrier, it does exhibit a mitigating effect on the damage.In HUVEC cells, a significant decrease in IL-8 levels was observed in sesamol and sesamol + CSE groups. In T98G cells, IL-8 levels were elevated in the CSE group, while a reduction was observed in the sesamol and sesamol + CSE groups. TNF-α levels went up in the CSE group but down in the sesamol and sesamol + CSE groups in T98G cells. Furthermore, the IL-6 levels were significantly increased in both the sesamol and sesamol + CSE groups in HUVEC cells, while a decrease was noted in T98G cells in sesamol treatment. The increase in IL-8 and TNF-α levels in T98G cells due to CSE indicates an inflammatory response. It can contribute to the enhanced BBB permeability. As a result, sesamol reduced inflammation caused by CSE by controlling IL-8, IL-6, and TNF-α. This molecule may serve a therapeutic role by diminishing inflammation and protecting the blood-brain barrier from damage.
Collapse
Affiliation(s)
- Ildem Koru
- Department of Biotechnology, Graduate School of Science, Uskudar University, Istanbul, Turkey
| | - Belkıs Atasever-Arslan
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, Turkey.
| |
Collapse
|
78
|
Mabrouk DM, El-Akad RH, Afifi AH, Sharaf HA, El-Sharkawy SL, El Makawy AI. In vivo and in silico studies on the potential role of garden cress oil in attenuating methotrexate-induced inflammation and apoptosis in liver. Sci Rep 2025; 15:6178. [PMID: 39979397 PMCID: PMC11842783 DOI: 10.1038/s41598-025-89550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025] Open
Abstract
Methotrexate (MTX) has been used in high doses for cancer therapy and low doses for autoimmune diseases. It is proven that methotrexate-induced hepatotoxicity occurs even at relatively low doses. It is known that garden cress has anti-inflammatory, antioxidant, and hepatoprotective properties. This study investigates the potential alleviating effect of garden cress oil (GCO) against MTX-induced hepatotoxicity in rats. The chemical composition of GCO was assessed using GC/MS analysis. Liver damage was studied using hepatotoxicity biomarkers, molecular, and histological analysis. Also, the effects of GCO on TNF-α and caspase-3 proteins were evaluated through molecular docking studies. The results demonstrated that MTX caused liver damage, as seen by elevated levels of the liver enzymes ALT, AST, and ALP. Likewise, MTX showed clear signs of apoptosis, such as increased mRNA expression levels of BAX, Caspase-3, and P53, and increased liver inflammation indicated by higher levels of TNF-α expression. MTX exhibited significant liver damage, as demonstrated by histological examination. Treatment with GCO effectively alleviated the apoptotic effects of MTX, provided protection against inflammation, and restored histological alterations. GC/MS metabolite profiling of garden cress oil revealed the presence of several phytoconstituents, including tocopherols, erucic acid, sesamolin, linoleic acid, vaccenic acid, oleic acid, stearic acid, and palmitic acid, that showed strong binding affinities toward TNF-α and caspase-3 proteins in molecular docking studies, which could explain the anti-apoptotic and anti-inflammatory potential of GCO.
Collapse
Affiliation(s)
- Dalia M Mabrouk
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, P.O.12622, Giza, Egypt
| | - Radwa H El-Akad
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, PO Box 12622, Cairo, Egypt
| | - Ahmed H Afifi
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, PO Box 12622, Cairo, Egypt
| | - Hafiza A Sharaf
- Pathology Department, Medical Research and Clinical Studies Institute, National Research Centre, P.O.12622, Giza, Egypt
| | - Sonia L El-Sharkawy
- Pathology Department, Medical Research and Clinical Studies Institute, National Research Centre, P.O.12622, Giza, Egypt
| | - Aida I El Makawy
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, P.O.12622, Giza, Egypt.
| |
Collapse
|
79
|
Ponce-Regalado MD, Becerril-Villanueva E, Maldonado-García JL, Moreno-Lafont MC, Martínez-Ramírez G, Jacinto-Gutiérrez S, Arreola R, Sánchez-Huerta K, Contis-Montes de Oca A, López-Martínez KM, Bautista-Rodríguez E, Chin-Chan JM, Pavón L, Pérez-Sánchez G. Comprehensive view of suicide: A neuro-immune-endocrine approach. World J Psychiatry 2025; 15:98484. [PMID: 39974471 PMCID: PMC11758041 DOI: 10.5498/wjp.v15.i2.98484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/26/2024] [Accepted: 12/23/2024] [Indexed: 01/14/2025] Open
Abstract
Suicide is defined as the act of a person attempting to take their own life by causing death. Suicide is a complex phenomenon that is influenced by a multitude of factors, including psychosocial, cultural, and religious aspects, as well as genetic, biochemical, and environmental factors. From a biochemical perspective, it is crucial to consider the communication between the endocrine, immune, and nervous systems when studying the etiology of suicide. Several pathologies involve the bidirectional communication between the peripheral activity and the central nervous system by the action of molecules such as cytokines, hormones, and neurotransmitters. These humoral signals, when present in optimal quantities, are responsible for maintaining physiological homeostasis, including mood states. Stress elevates the cortisol and proinflammatory cytokines levels and alter neurotransmitters balance, thereby increasing the risk of developing a psychiatric disorder and subsequently the risk of suicidal behavior. This review provides an integrative perspective about the neurochemical, immunological, and endocrinological disturbances associated with suicidal behavior, with a particular focus on those alterations that may serve as potential risk markers and/or indicators of the state preceding such a tragic act.
Collapse
Affiliation(s)
- María D Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Jalisco, Mexico
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Martha C Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Gabriela Martínez-Ramírez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
- Facultad de Medicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional autónoma de México, Tlalnepantla 54090, Mexico
| | - Salomón Jacinto-Gutiérrez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Rodrigo Arreola
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Karla Sánchez-Huerta
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico
| | - Arturo Contis-Montes de Oca
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | | | - José Miguel Chin-Chan
- Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Campeche, Campeche 24039, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| |
Collapse
|
80
|
Jiang J, Tian Y, Wu X, Zeng M, Wu C, Wei D, Luo H, Sun J, Ding J, Fan H. Temperature and light dual-responsive hydrogels for anti-inflammation and wound repair monitoring. J Mater Chem B 2025; 13:2855-2870. [PMID: 39882768 DOI: 10.1039/d4tb02555e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Wound healing is a complex and dynamic biological process that requires meticulous management to ensure optimal outcomes. Traditional wound dressings, such as gauze and bandages, although commonly used, often fall short in their frequent need for replacement, lack of real-time monitoring and absence of anti-inflammatory and antibacterial properties, which can lead to increased risk of infection and delayed healing. Here, we address these limitations by introducing an innovative hydrogel dressing, named PHDNN6, to combine wireless Bluetooth temperature monitoring and light-triggered nitric oxide (NO) release to enhance wound healing and management. The PHDNN6 hydrogel is based on a poly(N-isopropylacrylamide) (PNIPAM) matrix, integrated with methacrylated and dopamine-grafted hyaluronic acid (HA-MA-DA), which allows the dressing to be highly responsive to changes in wound temperature, enabling continuous and real-time monitoring of the wound microenvironment wirelessly. Besides, PHDNN6 is embedded with photothermal polydopamine nanoparticles (PDA NPs) that are loaded with a NO donor, N,N'-di-sec-butyl-N,N'-dinitroso-1,4-phenylenediamine (BNN6). When exposed to near-infrared (NIR) laser irradiation, these PDA@BNN6 nanoparticles release NO to provide potent antibacterial and anti-inflammatory effects. The integration of continuous wireless temperature monitoring with NO release within a single hydrogel dressing represents a significant advancement in clinical wound care. This dual-functional platform not only provides real-time diagnostic capabilities but also offers therapeutic interventions to manage wound infections and promote tissue regeneration. Our research highlights the potential of PHDNN6 to revolutionize wound management by offering a comprehensive solution that addresses both the diagnostic and therapeutic needs in wound healing.
Collapse
Affiliation(s)
- Ji Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Yuan Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Xiaoyang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Mingze Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, P. R. China
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Hongrong Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jie Ding
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| |
Collapse
|
81
|
Wu CJ. NEMO Family of Proteins as Polyubiquitin Receptors: Illustrating Non-Degradative Polyubiquitination's Roles in Health and Disease. Cells 2025; 14:304. [PMID: 39996775 PMCID: PMC11854354 DOI: 10.3390/cells14040304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
The IκB kinase (IKK) complex plays a central role in many signaling pathways that activate NF-κB, which turns on a battery of genes important for immune response, inflammation, and cancer development. Ubiquitination is one of the most prevalent post-translational modifications of proteins and is best known for targeting substrates for proteasomal degradation. The investigations of NF-κB signaling pathway primed the unveiling of the non-degradative roles of protein ubiquitination. The NF-κB-essential modulator (NEMO) is the IKK regulatory subunit that is essential for IKK activation by diverse intrinsic and extrinsic stimuli. The studies centered on NEMO as a polyubiquitin-binding protein have remarkably advanced understandings of how NEMO transmits signals to NF-κB activation and have laid a foundation for determining the molecular events demonstrating non-degradative ubiquitination as a major driving element in IKK activation. Furthermore, these studies have largely solved the enigma that IKK can be activated by diverse pathways that employ distinct sets of intermediaries in transmitting signals. NEMO and NEMO-related proteins that include optineurin, ABIN1, ABIN2, ABIN3, and CEP55, as non-degradative ubiquitin chain receptors, play a key role in sensing and transmitting ubiquitin signals embodied in different topologies of polyubiquitin chains for a variety of cellular processes and body responses. Studies of these multifaceted proteins in ubiquitin sensing have promoted understanding about the functions of non-degradative ubiquitination in intracellular signaling, protein trafficking, proteostasis, immune response, DNA damage response, and cell cycle control. In this review, I will also discuss how dysfunction in the NEMO family of protein-mediated non-degradative ubiquitin signaling is associated with various diseases, including immune disorders, neurodegenerative diseases, and cancer, and how microbial virulence factors target NEMO to induce pathogenesis or manipulate host response. A profound understanding of the molecular bases for non-degradative ubiquitin signaling will be valuable for developing tailored approaches for therapeutic purposes.
Collapse
Affiliation(s)
- Chuan-Jin Wu
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
82
|
Qin SJ, Zeng QG, Zeng HX, Meng WJ, Wu QZ, Lv Y, Dai J, Dong GH, Zeng XW. Novel perspective on particulate matter and Alzheimer's disease: Insights from adverse outcome pathway framework. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125601. [PMID: 39756567 DOI: 10.1016/j.envpol.2024.125601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, that accounts for 50-75% of all dementia cases. Evidence demonstrates the link between particulate matter (PM) exposure and AD. However, there are still considerable research gaps. This review aims to clarify the mechanism between PM and AD from different levels (subcellular/cellular/system/population) by using an adverse outcome pathway (AOP) framework. We applied a chemical-phenotype interaction network-based workflow to integrate diverse genes and phenotypes. The interactions among PM, genes, phenotypes, and AD were retrieved from the Comparative Toxicogenomics Database (CTD), DisGeNET, MalaCards, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG), which are publicly available databases. The filtered genes and phenotypes were assembled as molecular initiating events (MIEs) and key events (KEs) according to the upstream and downstream relationships, generating a predictive PM-Gene-Phenotype-AD AOP network. According to the Organization for Economic Co-operation and Development handbook (OECD), a verified AOP network was assessed and applied to determine the effects of PM on AD. PM could increase APP and GSK3B, increase apoptosis, impair cognition and memory, and ultimately lead to AD. Overall, chemical-phenotype interactions are expressed in a formal structured notation using controlled terms for chemicals, phenotypes, taxons, and anatomical descriptors. To our knowledge, this is the first AOP framework focusing on the underlying mechanism of exposure to PM on AD. Our network-based approach not only fills mechanism gaps in PM and AD but sheds light on constructing AOP frameworks for new chemicals.
Collapse
Affiliation(s)
- Shuang-Jian Qin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Guo Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui-Xian Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen-Jie Meng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Zhen Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan Lv
- Department of Neurology, Jiangbin Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Jian Dai
- Department of Clinical Psychology, Jiangbin Hospital, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Wen Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
83
|
Cousin A, Oger M, de Jenlis A, Lejart A, Barbier L, Riccobono D, Holy X, Favier AL, Nikovics K. CD163, a novel receptor for TNF, was revealed in situ by proximity ligation assay. Heliyon 2025; 11:e42194. [PMID: 39995928 PMCID: PMC11848091 DOI: 10.1016/j.heliyon.2025.e42194] [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: 07/27/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Cytokine therapy utilizes cytokines to enhance the immune system to fight diseases. These strategies rely on advanced knowledge, including the communication between cytokines and their receptors. In situ, cytokine-receptor interactions are typically analyzed by co-localization using immunolabeling. Our study compared co-localization using the Proximity Ligation Assay (PLA), a recently developed in situ protein-protein interaction technique. In an inflamed porcine lung model, we demonstrated the efficacy of PLA in detecting interactions between tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2. Additionally, the CD163 receptor was identified as a novel partner of TNF. Furthermore, the combination of immunolabeling and PLA offered additional insights, particularly, the internalization of TNF following its binding with CD163 in macrophages. Our work focused on in situ interactions of TNF with macrophages TNF receptors and suggested exciting perspectives for further understanding and application of cytokine-based therapies.
Collapse
Affiliation(s)
- Alexandre Cousin
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Myriam Oger
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Aymar de Jenlis
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Audrey Lejart
- Molecular Biology Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Laure Barbier
- Molecular Biology Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Diane Riccobono
- Radiobiology Unit, Department of NRBC Defense, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Xavier Holy
- Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Anne-Laure Favier
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| | - Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223, Brétigny-sur-Orge, France
| |
Collapse
|
84
|
Guo P, Xue Y, Zhang D, Lu Q, Liu Y, Xiong J, Ye C, Fu S, Wu Z, Wang X, Qiu Y. Network Pharmacology to Unveil the Mechanism of Berberine in the Treatment of Streptococcus suis Meningitis in Humans and Pigs. TOXICS 2025; 13:138. [PMID: 39997953 PMCID: PMC11860940 DOI: 10.3390/toxics13020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Streptococcus suis (S. suis) is a major swine pathogen throughout the world as well as an emerging zoonotic agent. Among the symptoms caused by S. suis, including septicemia, pneumonia, endo-carditis, arthritis, and meningitis, the latter is the most overlooked. In the present study, we explored the mechanism of action of berberine against S. suis meningitis by obtaining berberine-related action targets, porcine S. suis meningitis targets, and human S. suis meningitis targets from open databases. We constructed a protein-protein interaction (PPI) network by using the STRING database and employed Cytoscape 3.8.0 to screen for core targets. We performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses through DAVID. We identified 31 potential targets of berberine, of which Toll-like receptor 4 (TLR4), fibronectin 1 (FN1), superoxide dismutase (SOD1), and catalase (CAT) were the four most critical targets. GO analysis revealed the enrichment of terms related to the response to oxidative stress and the inflammatory response. KEGG analysis revealed the enrichment of the interleukin 17 (IL-17), phosphoinositide 3-kinase (PI3K)-Akt, TLR, tumor necrosis factor (TNF), and mitogen-activated protein kinase (MAPK) signaling pathways. In addition, the admetSAR results showed that berberine can cross the blood-brain barrier. The molecular docking results indicated key binding activity between TLR4-berberine and FN1-berberine. In summary, berberine protects against Streptococcus suis meningitis by regulating inflammatory response and oxidative stress in humans and pigs. Our study updates the current knowledge of the targets of S. suis meningitis to exploit new drugs in humans and pigs, to develop environmentally friendly and antibiotic-free animal-derived food products, and to improve the farming industry and economic development.
Collapse
Affiliation(s)
- Pu Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yunda Xue
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dan Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jianglin Xiong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (P.G.); (Y.X.); (D.Z.); (Q.L.); (Y.L.); (J.X.); (C.Y.); (S.F.); (Z.W.)
- Wuhan Engineering and Technology Research Center of Animal Disease-Resistant Nutrition, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| |
Collapse
|
85
|
Theodoridis D, Tsifi A, Magiorkinis E, Tsamakidis X, Voulgaridis A, Moustaferi E, Skrepetou N, Tsifis S, Ioannidis A, Chronopoulos E, Chatzipanagiotou S. The Role of Monocyte Distribution Width (MDW) in the Prediction of Death in Adult Patients with Sepsis. Microorganisms 2025; 13:427. [PMID: 40005792 PMCID: PMC11858437 DOI: 10.3390/microorganisms13020427] [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: 01/15/2025] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Sepsis is a life-threatening condition; it is a major cause of hospital mortality worldwide and it constitutes a global health problem. This research investigates the use of MDW as a predictor for septic patients. This was a double-center prospective cohort study of adult septic patients. Septic patients were identified and were categorized into two categories: those who improved and those who died. Blood was drawn from the patients three times, on the first, third, and fifth day of their admission to the hospital. MDW was evaluated as a biomarker to predict patient outcome. In addition, existing inflammatory markers were recorded in all patients. The MDW was able to predict patient's outcome. The average MDW was found to be significantly higher in patients who died in all records. For example, an MDW value of 28.4 on the first day of admission to the hospital was shown to be the best cut-off value in determining fatal outcomes; receiver operating characteristic (ROC) analysis revealed an area under the curve value of 0.71 (95% Confidence Interval-CI: 0.57-0.84) with a sensitivity of 64.7% and a specificity of 88.2%. In conclusion, MDW, in addition to being a marker that can quickly detect sepsis more effectively than other biomarkers, which is proven by numerous studies, could also be used as an indicator to predict patient outcome. This work is an attempt in that direction.
Collapse
Affiliation(s)
| | - Angeliki Tsifi
- Department of Pathophysiology, General Hospital of Athens LAIKO, 11527 Athens, Greece;
| | | | - Xenofon Tsamakidis
- Gastroenterology Clinic, Oncology Hospital of Athens “Saint Savvas”, 11522 Athens, Greece;
| | | | - Evgenia Moustaferi
- Hematology Laboratory, Konstantopoulio General Hospital, 14233 Nea Ionia, Greece;
| | - Nikoletta Skrepetou
- Hematology Department, Konstantopoulio General Hospital, 14233 Nea Ionia, Greece;
| | - Sotirios Tsifis
- Department of Infectious Diseases, Fondazione IRCCS Policlinico Sa Matteo, 27100 Pavia, Italy;
| | - Anastasios Ioannidis
- Laboratory of Basic Health Sciences, Department of Nursing, Faculty of Health Sciences, University of Peloponnese, 22100 Tripoli, Greece;
| | | | - Stylianos Chatzipanagiotou
- Department of Biopathology and Clinical Microbiology, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| |
Collapse
|
86
|
Qin Y, Chen X, Bao L, Ren L, Dou G, Lian J, Xing S, Li Z, Ding F, Qin W, Liu X, Zhu B, Liu S, Jin Z, Yang X. Lipid metabolism of apoptotic vesicles accelerates cutaneous wound healing by modulating macrophage function. J Nanobiotechnology 2025; 23:106. [PMID: 39939963 PMCID: PMC11823102 DOI: 10.1186/s12951-025-03194-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 02/01/2025] [Indexed: 02/14/2025] Open
Abstract
The application of apoptotic extracellular vesicles (ApoEVs) derived from stem cell in skin wound healing has garnered significant attention. In recent decades, scholars have shown that extracellular vesicles (EVs) established intercellular communication by carrying proteins or microRNAs, the role of lipids in EVs in wound healing has yet to be clarified. Here, we focus on the key role of group X secretory phospholipase A2 (sPLA2-X) in lipid metabolism. Specifically, sPLA2-X significantly increased the production of the anti-inflammatory lipid mediators, resolvin D5 (RvD5), by hydrolyzing phospholipids in ApoEVs. This change not only promoted the uptake of ApoEVs by macrophages, but also effectively inhibited the expression of tumor necrosis factor-alpha (TNF-α) in macrophages, promoting the healing of skin wounds. In summary, this study contributes to our understanding of the mechanisms by which ApoEVs support skin defect repair and offers a potential theoretical approach for using ApoEVs in skin wound treatment. With further research and optimization, it is expected that more efficient and secure ApoEVs-based treatment strategies will be developed, bringing new breakthroughs in clinical treatment of skin injuries and related diseases.
Collapse
Affiliation(s)
- Yuan Qin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Disease, School of Stomatology, The Fourth Military Medical University, Xi' an, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Lili Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Disease, School of Stomatology, The Fourth Military Medical University, Xi' an, 710032, China
| | - Lili Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Geng Dou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jianing Lian
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Shujuan Xing
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
- College of Life Science, Northwest University, Xi'an, 710069, China
| | - Zihan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Feng Ding
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wen Qin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xulin Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Zhu
- Outpatient Department, General Hospital of Xizang Military Region, Lhasa, 850007, China
| | - Shiyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Disease, School of Stomatology, The Fourth Military Medical University, Xi' an, 710032, China.
| | - Zuolin Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Xiaoshan Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology & Clinic of Oral Rare Diseases and Genetic Disease, School of Stomatology, The Fourth Military Medical University, Xi' an, 710032, China.
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
87
|
Simond A, Lesage V, Verreault J, Loseto L, Houde M, Elliott A, Noël M, Brown TM. Contaminant-Associated Disruption of the Skin Transcriptome in the Endangered St. Lawrence Estuary Beluga. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:2389-2399. [PMID: 39874536 PMCID: PMC11823453 DOI: 10.1021/acs.est.4c08272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/30/2025]
Abstract
The St. Lawrence Estuary (SLE) beluga (Delphinapterus leucas) population in Canada is Endangered, and endocrine disrupting contaminants, such as polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs), and other halogenated flame retardants, have been identified as a threat to the recovery of this population. Here, potential impacts of these contaminants on SLE beluga were evaluated by comparing skin transcriptome profiles and biological pathways between this population and a population less exposed to contaminants (Eastern Beaufort Sea) used as a reference. Differential gene expression analysis indicated potential seasonal or geography-related (Arctic vs temperate regions) effects on the skin transcriptome. Among the gene transcripts that were associated with Σ31PCB (123 genes), Σ29PBDE (198 genes), HBB (347 genes), and PBEB (126 genes) blubber concentrations, several were related to immune response pathways. In addition, 18 toxicology-related gene transcripts selected from the literature were correlated with organohalogen concentrations and were used to derive new threshold values in beluga skin for potential biological effects of ΣPCB (1,500 ng/g lw), ΣPBDE (52 ng/g lw), and two other flame retardants, i.e., HBB (1.2 ng/g lw) and PBEB (0.04 ng/g lw). Results suggest that exposure to these organohalogens pose a risk to the immune system of SLE beluga.
Collapse
Affiliation(s)
- Antoine
É. Simond
- Fisheries
and Oceans Canada, Pacific Science Enterprise
Center, 4160 Marine Drive, West Vancouver, British Columbia V7V 1N6, Canada
- Department
of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Véronique Lesage
- Institut
Maurice Lamontagne, Fisheries and Oceans
Canada, Mont-Joli, Quebec G5H 3Z4, Canada
| | - Jonathan Verreault
- Centre
de recherche en toxicologie de l’environnement (TOXEN), Département
des sciences biologiques, Université
du Québec à Montréal, P.O. Box 8888, Succursale centre-ville, Montreal, Quebec H3C 3P8, Canada
| | - Lisa Loseto
- Freshwater
Institute, Fisheries and Oceans Canada, 501 University Crescent, Winnipeg, Manitoba R3T 2N6, Canada
- Centre for
Earth Observation Science, University of
Manitoba, 535 Wallace
Building, 125 Dysart Road, Winnipeg, Manitoba R3T 2N2, Canada
| | - Magali Houde
- Environment
and Climate Change Canada, Centre Saint-Laurent, 105 McGill Street, Montreal, Quebec H2Y 2E7, Canada
| | - Ashley Elliott
- Freshwater
Institute, Fisheries and Oceans Canada, 501 University Crescent, Winnipeg, Manitoba R3T 2N6, Canada
| | - Marie Noël
- Freshwater
Institute, Fisheries and Oceans Canada, 501 University Crescent, Winnipeg, Manitoba R3T 2N6, Canada
| | - Tanya M. Brown
- Fisheries
and Oceans Canada, Pacific Science Enterprise
Center, 4160 Marine Drive, West Vancouver, British Columbia V7V 1N6, Canada
| |
Collapse
|
88
|
Wang Q, Qin B, Yu H, Zeng J, Fan J, Wu Q, Zeng R, Yu H, Zhang X, Li M, Zhou Y, Diao L. Mitigating effects of Jiawei Chaihu Shugan decoction on necroptosis and inflammation of hippocampal neurons in epileptic mice. Sci Rep 2025; 15:4649. [PMID: 39920301 PMCID: PMC11805973 DOI: 10.1038/s41598-025-89275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/04/2025] [Indexed: 02/09/2025] Open
Abstract
Jiawei Chaihu Shugan decoction (JWCHSGD) is a traditional Chinese medicine well-known for its beneficial effects in treating epilepsy (Xianzheng in ancient Chinese), but the molecular mechanism of its action remains unclear. To investigate the molecular mechanism of JWCHSGD's prevention of epilepsy-mediated neuron from necroptosis and inflammation via the circRNA-Csnk1g3/Csnk1g3-85aa/ CK1γ3/TNF-α signal pathway. In vitro, murine neuronal HT22 cells were treated in six groups: control, model, carbamazepine, and three JWCHSGD doses (high, medium, low). Viability and apoptosis were assessed via CCK-8 and flow cytometry. In vivo, 60 C57BL/6J mice were divided into six groups: control, model, carbamazepine, JWCHSGD, JWCHSGD + Sh Circ_Csnk1g3, and JWCHSGD + Sh NC. An epilepsy model was induced, and treatments were administered for two weeks. Outcomes included EEG, hippocampal histopathology, apoptosis (TUNEL), and mRNA/protein expression of key pathway markers. In HT22 cells, the model group showed reduced viability, increased apoptosis, and elevated mRNA/protein levels of Csnk1g3-85aa, RIP1, RIP3, MLKL, TNF-α, IL-6, and IL-1β (P < 0.05). JWCHSGD and carbamazepine increased viability and decreased apoptosis, reversing these molecular changes (P < 0.05). In mice, the model group had heightened epileptic discharges, neuronal damage, and apoptosis, along with increased expression of the same markers (P < 0.05). JWCHSGD and carbamazepine mitigated these effects (P < 0.05). JWCHSGD reduces epileptic events by regulating the circRNA-Csnk1g3/Csnk1g3-85aa/CK1γ3/TNF-α signaling pathway, impacting necroptosis and inflammation in hippocampal neurons and HT22 cells.
Collapse
Affiliation(s)
- Qin Wang
- The First Clinical School of Medicine, Guangxi University of Chinese Medicine, 179 Mingxiu East Road, Nanning, 530001, Guangxi, China
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Qingxiu District, Nanning, 530023, Guangxi, China
| | - Baijun Qin
- Department of Gastroenterology, Chongqing City Hospital of Traditional Chinese Medicine, No. 6, Panxi seventh branch road, Jiangbei District, Chongqing, 400021, China
| | - Han Yu
- Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jiawei Zeng
- The First Clinical School of Medicine, Guangxi University of Chinese Medicine, 179 Mingxiu East Road, Nanning, 530001, Guangxi, China
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Qingxiu District, Nanning, 530023, Guangxi, China
| | - Jingjing Fan
- The First Clinical School of Medicine, Guangxi University of Chinese Medicine, 179 Mingxiu East Road, Nanning, 530001, Guangxi, China
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Qingxiu District, Nanning, 530023, Guangxi, China
| | - Qiong Wu
- Xinyang Central Hospital, Xinyang, 464000, Henan, China
| | - Rong Zeng
- Qinzhou Maternal and Child Health Hospital (Qinzhou Red Cross Hospital), No.1 Anzhou Avenue, Qinzhou City, Guangxi Zhuang Autonomous Region, China
| | - Haichun Yu
- Guangxi Technological College of Machinery and Electricity, Nanning, 530007, Guangxi, China
| | - Xian Zhang
- Guangxi Zhuang Autonomous Region Brain Hospital, Liuzhou, 545005, Guangxi, China
| | - Mingfen Li
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Qingxiu District, Nanning, 530023, Guangxi, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, Guangxi, China
| | - Yanying Zhou
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Qingxiu District, Nanning, 530023, Guangxi, China
| | - Limei Diao
- The First Clinical School of Medicine, Guangxi University of Chinese Medicine, 179 Mingxiu East Road, Nanning, 530001, Guangxi, China.
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Qingxiu District, Nanning, 530023, Guangxi, China.
- Guangxi Zhuang Autonomous Region Brain Hospital, Liuzhou, 545005, Guangxi, China.
| |
Collapse
|
89
|
Chi Y, Zhang Y, Lin H, Zhou S, Jia G, Wen W. The association of lipid accumulation product with inflammatory parameters and mortality: evidence from a large population-based study. FRONTIERS IN EPIDEMIOLOGY 2025; 4:1503261. [PMID: 39967714 PMCID: PMC11832662 DOI: 10.3389/fepid.2024.1503261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/27/2024] [Indexed: 02/20/2025]
Abstract
Background Obesity is closely associated with lipid metabolism, and the accumulation of lipids leads to low-level inflammation in the body, which can trigger cardiovascular disease. This study aimed to explore the association between a novel marker of lipid accumulation, the abdominal volume index (AVI), inflammatory parameters, and mortality. Methods This study enrolled 2,109 older adult senior citizens (aged over 60 years) with hypertension from the National Health and Nutrition Examination Survey. The primary endpoints included all-cause mortality and cardiovascular mortality, which were assessed by linking the data to the National Death Index records. Cox regression model and subgroup analysis were constructed to investigate the associations between AVI and both all-cause and cardiovascular mortality. Restricted cubic splines were employed to further explore the relationships among AVI, inflammatory parameters, and mortality. By considering inflammatory factors as mediators, we investigate the mediating effects of AVI on mortality. Results After a median follow-up of 69 months, there were 1,260 deaths, with 337 attributed to cardiovascular causes within the older adult population studied. In the multivariable-adjusted model, AVI was positively associated with both all-cause and cardiovascular mortality [Hazard Ratio (HR) = 1.09, 95% CI = 1.06-1.11 for all-cause mortality; HR = 1.07, 95% CI = 1.03-1.12 for cardiovascular mortality]. Kaplan-Meier survival plots indicated an overall median survival time of 144 months. Mediation analysis revealed that Systemic Inflammatory Response Index (SIRI), Monocyte-to-HDL ratio (MHR), and Neutrophil-to-Lymphocyte ratio (NLR) mediated 27.15%, 35.15%, and 16.55%, respectively, of the association between AVI and all-cause mortality. Conclusion AVI is positively associated with all-cause mortality in older adults with hypertension, and this association appears to be partially mediated by inflammatory parameters.
Collapse
Affiliation(s)
- Yi Chi
- Department of Integrative Medicine (Geriatrics), The People’s Hospital Medical Group of Xiangzhou, Zhuhai, China
| | - Yiqing Zhang
- Department of Integrative Medicine (Geriatrics), The People’s Hospital Medical Group of Xiangzhou, Zhuhai, China
| | - Huang Lin
- Department of Integrative Medicine (Geriatrics), The People’s Hospital Medical Group of Xiangzhou, Zhuhai, China
| | - Shanshan Zhou
- Department of Integrative Medicine (Geriatrics), The People’s Hospital Medical Group of Xiangzhou, Zhuhai, China
| | - Genlin Jia
- Department of Spleen and Gastroenteritis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Wen
- Department of Cardiovascular Disease, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
90
|
Hajikhezri Z, Zygouras I, Sönnerborg A, van Domselaar R. Pan-caspase inhibitors induce secretion of HIV-1 latency reversal agent lymphotoxin-alpha from cytokine-primed NK cells. Cell Death Discov 2025; 11:44. [PMID: 39905001 PMCID: PMC11794648 DOI: 10.1038/s41420-025-02330-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
The persistence of HIV-1 latency reservoirs in CD4+ T cells is a significant obstacle for curing HIV-1. Shock-and-kill strategies, which aim to reactivate latent HIV-1 followed by cytotoxic clearance, have shown limited success in vivo due to insufficient efficacy of latency reversal agents (LRAs) and off-target effects. Natural killer (NK) cells, with their ability to mediate cytotoxicity independent of antigen specificity, offer a promising avenue for enhancing the shock-and-kill approach. Previously, we observed that pan-caspase inhibitors induce NK cells to secrete an LRA in vitro. Here, we aimed to identify this LRA using a targeted proteomic approach. We identified lymphotoxin-α (LTα) as the key LRA secreted by NK cells following pan-caspase inhibitor treatment. LTα was shown to significantly induce HIV-1 LTR promoter activity, a hallmark of viral reactivation. Neutralization of LTα effectively abolished the observed LRA activity, confirming its central role. Moreover, cytokine-primed but not resting human primary NK cells exhibited LRA activity that could be neutralized with LTα neutralizing antibodies. Finally, pan-caspase inhibitor treatment did not decrease the ability of the cytokine-primed NK cells to kill target cells. These findings demonstrate that cytokine-primed NK cells, through LTα secretion, can effectively reactivate latent HIV-1 following pan-caspase inhibitor treatment, without compromising NK cell cytotoxicity. This highlights a potential enhancement strategy utilizing NK cells for shock-and-kill approaches in HIV-1 cure research.
Collapse
Affiliation(s)
- Zamaneh Hajikhezri
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Zygouras
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Microbiology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Robert van Domselaar
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
91
|
Rashid K, Kalthoff H, Abdulkadir SA, Adam D. Death ligand receptor (DLR) signaling: Its non-apoptotic functions in cancer and the consequences of DLR-directed therapies. Drug Discov Today 2025; 30:104299. [PMID: 39842503 DOI: 10.1016/j.drudis.2025.104299] [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: 09/27/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Death ligands (DLs), particularly tumor necrosis factor alpha (TNF-α), FAS ligand (FASL), and TNF-related apoptosis-inducing ligand (TRAIL), collectively termed TFT, are pivotal members of the TNF superfamily. While traditionally linked to apoptosis, TFT proteins have emerged as key regulators of various non-apoptotic processes. This review summarizes the non-apoptotic functions of TFT in cancer and explores the intricate crosstalk signaling pathways and their impact on nuclear factor kappa B (NF-κB) signaling, inflammation, and pro-tumorigenic function. It also highlights the potential connections and hurdles that exist in translating synthetic lethality strategies involving DLs into clinical applications. Lastly, it discusses the challenges and opportunities associated with TFT-targeted therapeutic strategies for both malignant and non-malignant diseases.
Collapse
Affiliation(s)
- Khalid Rashid
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Kiel University (CAU), Kiel, Germany
| | - Sarki A Abdulkadir
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dieter Adam
- Institute of Immunology, Kiel University (CAU), Kiel, Germany
| |
Collapse
|
92
|
Patel TR, Welch CM. The Science of Cholesteatoma. Otolaryngol Clin North Am 2025; 58:1-27. [PMID: 39353746 DOI: 10.1016/j.otc.2024.07.022] [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: 10/04/2024]
Abstract
Cholesteatoma is a potential end-stage outcome of chronic ear infections that can result in the destruction of temporal bone structures with potential resultant hearing loss, vertigo, and intracranial infectious complications. There is currently no treatment apart from surgery for this condition, and despite years of study, the histopathogenesis of this disease remains poorly understood. This review is intended to summarize our accumulated knowledge of the mechanisms of cholesteatoma development and the underlying molecular biology. Attention will be directed particularly to recent developments, covering many potential pharmacologic targets that could be used to treat this disease in the future.
Collapse
Affiliation(s)
- Tirth R Patel
- Division of Otology/Neurotology-Skull Base Surgery, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, 1500 Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Christopher M Welch
- Division of Otology/Neurotology-Skull Base Surgery, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, 1500 Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
93
|
Su Y, Huang M, Chen Q, He J, Li S, Wang M. Harnessing β-glucan conjugated quercetin nanocomplex to function as a promising anti-inflammatory agent via macrophage-targeted delivery. Carbohydr Polym 2025; 349:122952. [PMID: 39638531 DOI: 10.1016/j.carbpol.2024.122952] [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: 07/04/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Quercetin, a promising anti-inflammatory agent, faces challenges related to poor bioavailability and limited practical applications. β-glucan, a natural polysaccharide, can be specifically recognized by macrophages, making it an ideal targeting carrier to enhance therapeutic efficacy for macrophage-related dysfunctions. In this study, β-glucan conjugated quercetin nano-complexes (CM-Cur@QT) were developed to target macrophage and alleviate pro-inflammatory response in M1-like macrophages. The results demonstrated that CM-Cur@QT exhibited a spheric shape with an average diameter around 200 nm. FT-IR, 1H NMR, XRD and XPS analyses confirmed the complexation of CM-Cur@QT. This complex showed excellent stability during stimulated digestion, protecting QT from degradation while maintaining favorable antioxidant activity. After complexation, CM-Cur@QT displayed sustained uptake kinetics and enhanced accumulation in macrophages, with a 61.88 % increase compared to individual quercetin after 5 h of incubation. Meanwhile, CM-Cur@QT administration induced evidently cell cycle phases transitions and altered phagocytotic activity in M1-like macrophages. Furthermore, CM-Cur@QT reduced intracellular ROS accumulation, achieving a ROS scavenging rate of up to 49.92 %, compared to 25.59 % in quercetin group. This complex also effectively modulated TNF-a, IL-6 and TGF-β secretion profiles in pro-inflammatory macrophages, outperforming individual QT treatment. Notably, CM-Cur@QT facilitated anti-inflammatory effects while minimizing impacts on inactivated M0 macrophages. These findings underscore the potential of CM-Cur@QT as a promising agent for mitigating inflammatory disorders.
Collapse
Affiliation(s)
- Yuting Su
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Manting Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Qiaochun Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Jiayi He
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Siqian Li
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
94
|
Bosáková V, Papatheodorou I, Kafka F, Tomášiková Z, Kolovos P, Hortová Kohoutková M, Frič J. Serotonin attenuates tumor necrosis factor-induced intestinal inflammation by interacting with human mucosal tissue. Exp Mol Med 2025; 57:364-378. [PMID: 39894823 PMCID: PMC11873120 DOI: 10.1038/s12276-025-01397-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/13/2024] [Accepted: 11/19/2024] [Indexed: 02/04/2025] Open
Abstract
The intestine hosts the largest immune system and peripheral nervous system in the human body. The gut‒brain axis orchestrates communication between the central and enteric nervous systems, playing a pivotal role in regulating overall body function and intestinal homeostasis. Here, using a human three-dimensional in vitro culture model, we investigated the effects of serotonin, a neuromodulator produced in the gut, on immune cell and intestinal tissue interactions. Serotonin attenuated the tumor necrosis factor-induced proinflammatory response, mostly by affecting the expression of chemokines. Serotonin affected the phenotype and distribution of tissue-migrating monocytes, without direct contact with the cells, by remodeling the intestinal tissue. Collectively, our results show that serotonin plays a crucial role in communication among gut-brain axis components and regulates monocyte migration and plasticity, thereby contributing to gut homeostasis and the progression of inflammation. In vivo studies focused on the role of neuromodulators in gut inflammation have shown controversial results, highlighting the importance of human experimental models. Moreover, our results emphasize the importance of human health research in human cell-based models and suggest that the serotonin signaling pathway is a new therapeutic target for inflammatory bowel disease.
Collapse
Affiliation(s)
- Veronika Bosáková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ioanna Papatheodorou
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Filip Kafka
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Tomášiková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Marcela Hortová Kohoutková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.
| |
Collapse
|
95
|
Yadav S, Prasannan A, Venkatachalam K, Binesh A. Exploring the mechanism and crosstalk between IL-6 and IL- 1β on M2 macrophages under metabolic stress conditions. Cytokine 2025; 186:156852. [PMID: 39765025 DOI: 10.1016/j.cyto.2024.156852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/14/2025]
Abstract
Macrophages are highly variable immune cells that are important in controlling inflammation and maintaining tissue balance. The ability to polarize into two major types-M1, promoting inflammation, and M2, resolving inflammation and contributing to tissue repair-determines their specific roles in health and disease. M2 macrophages are particularly important for reducing inflammation and promoting tissue regeneration, but their function is shaped mainly by surrounding cells. This is evident in obesity, diabetes, and chronic inflammation. Although many cytokines regulate macrophage polarization, interleukin-6 (IL-6) and interleukin-1β (IL-1β) are major players, but their effects on M2 macrophage behavior under metabolic stress remain unclear. This study describes the intricacies within M2 macrophages concerning IL-6 and IL-1β signaling when under metabolic stress. Though, more frequently than not, IL-6 is labelled as pro-inflammatory, it can also behave as an anti-inflammatory mediator. On the other hand, IL-1β is the main pro-inflammatory agent, particularly in metabolic disorders. The relationship between these cytokines and the macrophages is mediated through important pathways such as JAK/STAT and NFκB, which get perturbed by metabolic stress. Therefore, metabolic stress also alters the functional parameters of macrophages, including alterations in mitochondrial metabolism, glycolytic and oxidative metabolism. Phosphorylation alters the kinetics involved in energy consumption and affects their polarization and their function. However, it has been suggested that IL-6 and IL-1β may work in concert or competition when inducing M2 polarization and, importantly, implicate cytokine release, phagocytic activity, and tissue repair processes. In this review, we discuss the recent literature on the participation of IL-6 and IL-1β cytokines in macrophage polarization and how metabolic stress changes cytokine functions and synergistic relations. A better understanding of these cytokines would serve as an important step toward exploring alternative antiviral strategies directed against metabolic disturbance and, hence, approve further endeavors.
Collapse
Affiliation(s)
- Shawna Yadav
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Anusha Prasannan
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Ambika Binesh
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India.
| |
Collapse
|
96
|
Tran NM, Truong AT, Nguyen DT, Dang TT. Profiling Pro-Inflammatory Proteases as Biomolecular Signatures of Material-Induced Subcutaneous Host Response in Immuno-Competent Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2309709. [PMID: 39630111 PMCID: PMC11792001 DOI: 10.1002/advs.202309709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 09/19/2024] [Indexed: 02/05/2025]
Abstract
Proteases are important modulators of inflammation, but they remain understudied in material-induced immune response, which is critical to clinical success of biomedical implants. Herein, molecular expression and proteolytic activity of three distinct proteases, namely neutrophil elastase, matrix metalloproteinases, cysteine cathepsins (cathepsin-K and cathepsin-B) are comprehensively profiled, in the subcutaneous host response of immuno-competent mice against different biomaterial implants. Quantitative non-invasive monitoring with activatable fluorescent probes reveals that different microparticulate materials induce distinct levels of protease activity with degradable poly(lactic-co-glycolic) acid inducing the strongest signal compared to nondegradable materials such as polystyrene and silica oxide. Furthermore, protein expression of selected proteases, attributable to both their inactive and active forms, notably deviates from their activities associated only with their active forms. Protease activity exhibits positive correlations with protein expression of pro-inflammatory cytokines tumor necrosis factor α and interleukin 6 but negative correlation with pro-fibrotic cytokine transforming growth factor β1. This study also demonstrates the predictive utility of protease activity as a non-invasive, pro-inflammatory parameter for evaluation of the anti-inflammatory effects of model bioactive compounds on material-induced host response. Overall, the findings provide new insights into protease presence in material-induced immune responses, facilitating future biomaterial assessment to evoke appropriate host responses for implant applications.
Collapse
Affiliation(s)
- Nam M.P. Tran
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| | - Anh T.H. Truong
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| | - Dang T. Nguyen
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| | - Tram T. Dang
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| |
Collapse
|
97
|
Choudhary R, Kumar P, Shukla SK, Bhagat A, Anal JMH, Kour G, Ahmed Z. Synthesis and potential anti-inflammatory response of indole and amide derivatives of ursolic acid in LPS-induced RAW 264.7 cells and systemic inflammation mice model: Insights into iNOS, COX2 and NF-κB. Bioorg Chem 2025; 155:108091. [PMID: 39755101 DOI: 10.1016/j.bioorg.2024.108091] [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: 12/13/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
Ursolic acid (3-hydroxy-urs-12-ene-28-oic acid, UA) is a pentacyclic triterpene present in numerous plants, fruits and herbs and exhibits various pharmacological effects. However, UA has limited clinical applicability since it is classified as BCS class IV molecule, characterized by low solubility, low oral bioavailability and low permeability. In the present study, UA was isolated from the biomass marc of Lavandula angustifolia and was structurally modified by an induction of indole ring at the C-3 position and amide group at the C-17 position with the aim to enhance its pharmacological potential. This modification resulted in the synthesis of a series of compounds which were investigated for their anti-inflammatory potential both in-vitro and in animal models in comparison to UA. In RAW 264.7 cells, UA and its derivatives were non-cytotoxic up to 10 µM. The derivative UA-1 exhibited a significantly lower IC50 (2.2 ± 0.4 µM) for NO inhibition compared to UA (17.5 ± 2.0 µM). Molecular docking showed strong interactions of UA-1 with TNF-α and NF-κB. UA-1 significantly reduced LPS-induced pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) in RAW 264.7 macrophages with the inhibition levels of 74.2 ± 2.1 % for TNF-α, 55.9 ± 3.7 % for IL-6 and 59.7 ± 4.2 % for IL-1β at 5.0 µM, respectively and reactive oxygen species while upregulating anti-inflammatory cytokine, IL-10. It also downregulated iNOS, COX-2, p-NF-κB p65, and p-IκBα at both mRNA and protein levels. In LPS-induced systemic inflammation mice model, UA-1 significantly lowered NO, TNF-α, IL-6, IL-1β and serum biochemical parameters, reduced tissue damage, and exhibited improved aqueous solubility and moderate lipophilicity. Overall, UA-1 demonstrated superior anti-inflammatory potential, improved solubility, and better therapeutic potential compared to UA.
Collapse
Affiliation(s)
- Rupali Choudhary
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Puneet Kumar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sanket K Shukla
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Asha Bhagat
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Jasha Momo H Anal
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Gurleen Kour
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Zabeer Ahmed
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
98
|
Engel K, Kulow VA, Chernyakov D, Willscher E, Fähling M, Edemir B. Segment specific loss of NFAT5 function in the kidneys is sufficient to induce a global kidney injury like phenotype. FASEB J 2025; 39:e70352. [PMID: 39874047 PMCID: PMC11774485 DOI: 10.1096/fj.202402497r] [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: 10/18/2024] [Revised: 12/17/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Nuclear factor of activated T-cells 5 (NFAT5) is a transcription factor known for its role in osmotic stress adaptation in the renal inner medulla, due to the osmotic gradient that is generated between the renal cortex and renal inner medulla. However, its broader implications in kidney injury and chronic kidney disease (CKD) are less understood. Here we used two different Cre deleter mice (Ksp1.3-Cre and Aqp2-Cre) to generate tubule segment and even cell type-specific NFAT5-deficient mice and performed extensive gene expression profiling. In both Nfat5 knockout models, we observed massive changes in gene expression pattern, with heightened inflammatory responses and renal injury, culminating in renal fibrosis. Interestingly, inflammatory responses were much more prominent in the Aqp2Cre+/-Nfat5fl/fl mice that lack NFAT5 only in the collecting duct. By analyzing gene expression in the medullary and cortical regions of the kidney separately, we confirmed that the loss of NFAT5 results in kidney injury that extends beyond hypertonic areas. Renal injury correlates with the expression level of genes involved in inflammatory response, injury severity, and cytokine signaling. Thus, NFAT5 is essential not only for adapting to osmotic stress but also for its loss of function, which induces activation of inflammatory response and cytokine signaling that might affect regions with functional NFAT5 expression.
Collapse
Affiliation(s)
- Kristina Engel
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Vera Anna Kulow
- Institute of Translational PhysiologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Dmitry Chernyakov
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Edith Willscher
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Michael Fähling
- Institute of Translational PhysiologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Bayram Edemir
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
- Institute for Physiology and Pathophysiology, Zentrum für Biomedizinische Ausbildung und Forschung (ZBAF)Witten/Herdecke UniversityWittenGermany
| |
Collapse
|
99
|
Mao J, Lei H, Xu P, Liu S, Zhou J, Mei M, Wang N, Zhang X. Identifying key components from Melastoma dodecandrum in TNF-α-induced osteoblast injury model through a combination of cell membrane chromatography and mass spectrometry. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118836. [PMID: 39326812 DOI: 10.1016/j.jep.2024.118836] [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: 04/06/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Melastoma dodecandrum (MD), a traditional ethnomedicine, has been widely used for the treatment of fractures, osteoarthritis, and osteoporosis due to its remarkable anti-inflammatory activity. However, the specific active components responsible for its therapeutic effects on orthopedic conditions remain unidentified. AIM OF THE STUDY This study aimed to screen and identify key active components in MD using a combination of cell membrane chromatography and mass spectrometry, followed by cellular validation. MATERIALS AND METHODS A TNF-α-induced osteoblast injury model and an osteoblast membrane chromatography screening system were established to select and identify chemical components of MD that directly act on osteoblasts. The protective effects of MD on osteoblasts were assessed by evaluating cell viability, alkaline phosphatase (ALP) activity, cell mineralization and the expression of osteogenesis-related proteins OCN, RUNX2, and the TNF-α receptor protein TNFR1. Validation of the activity of individual components was also conducted. RESULTS MD significantly improved the viability of osteoblasts under TNF-α-induced injury, enhanced ALP activity, stimulated the expression of OCN and RUNX2 proteins, and decreased the expression of TNFR1. Cell membrane chromatography screening identified 32 chemical components, including 21 flavonoids, 6 organic acids, 2 phenylpropanoids, 2 terpenes, and 1 nucleotide. Molecular docking revealed that isovitexin could bind to the specific receptor TNFR1 on the cell membrane. Furthermore, cellular validation demonstrated that isovitexin significantly protected osteoblasts. CONCLUSIONS MD and its pharmacologically active component, isovitexin, exhibit protective effects against TNF-α-induced inflammatory injury in osteoblasts, laying a solid foundation for future drug development.
Collapse
Affiliation(s)
- Jiale Mao
- Lishui TCM Hospital Affiliated to Zhejiang Chinese Medical University (Lishui Hospital of Traditional Chinese Medicine), Lishui, 323000, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Zhejiang Provincial Key Laboratory of She Medicine Inheritance, Innovation, Development and Application of Traditional Chinese Medicine, China; Lishui She Medicine Inheritance, Innovation, Development and Application Key Laboratory of Traditional Chinese Medicine, China.
| | - Houxing Lei
- Lishui TCM Hospital Affiliated to Zhejiang Chinese Medical University (Lishui Hospital of Traditional Chinese Medicine), Lishui, 323000, China; Zhejiang Provincial Key Laboratory of She Medicine Inheritance, Innovation, Development and Application of Traditional Chinese Medicine, China; Lishui She Medicine Inheritance, Innovation, Development and Application Key Laboratory of Traditional Chinese Medicine, China.
| | - Pingcui Xu
- Zhejiang Academy of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Hangzhou, 310005, China.
| | - Shuang Liu
- Lishui TCM Hospital Affiliated to Zhejiang Chinese Medical University (Lishui Hospital of Traditional Chinese Medicine), Lishui, 323000, China; Zhejiang Provincial Key Laboratory of She Medicine Inheritance, Innovation, Development and Application of Traditional Chinese Medicine, China; Lishui She Medicine Inheritance, Innovation, Development and Application Key Laboratory of Traditional Chinese Medicine, China.
| | - Jiwang Zhou
- Lishui TCM Hospital Affiliated to Zhejiang Chinese Medical University (Lishui Hospital of Traditional Chinese Medicine), Lishui, 323000, China.
| | - Mingrong Mei
- Zhejiang Provincial Ethnic Hospital, Jingning, 323500, China.
| | - Nani Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Zhejiang Academy of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Hangzhou, 310005, China.
| | - Xiaoqin Zhang
- Lishui TCM Hospital Affiliated to Zhejiang Chinese Medical University (Lishui Hospital of Traditional Chinese Medicine), Lishui, 323000, China; Zhejiang Provincial Key Laboratory of She Medicine Inheritance, Innovation, Development and Application of Traditional Chinese Medicine, China; Lishui She Medicine Inheritance, Innovation, Development and Application Key Laboratory of Traditional Chinese Medicine, China.
| |
Collapse
|
100
|
Ramón-Luing LA, Martínez-Gómez LE, Martinez-Armenta C, Martínez-Nava GA, Medina-Quero K, Pérez-Rubio G, Falfán-Valencia R, Buendia-Roldan I, Flores-Gonzalez J, Ocaña-Guzmán R, Selman M, López-Reyes A, Chavez-Galan L. TNF/IFN-γ Co-Signaling Induces Differential Cellular Activation in COVID-19 Patients: Implications for Patient Outcomes. Int J Mol Sci 2025; 26:1139. [PMID: 39940907 PMCID: PMC11817726 DOI: 10.3390/ijms26031139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
TNF and IFN-γ are key proinflammatory cytokines implicated in the pathophysiology of COVID-19. Toll-like receptor (TLR)7 and TLR8 are known to recognize SARS-CoV-2 and induce TNF and IFN-γ production. However, it is unclear whether TNF and IFN-γ levels are altered through TLR-dependent pathways and whether these pathways mediate disease severity during COVID-19. This study aimed to investigate the association between TNF/IFN-γ levels and immune cell activation to understand their role in disease severity better. We enrolled 150 COVID-19 patients, who were classified by their systemic TNF and IFN-γ levels (high (H) or normal-low (N-L)) as TNFHIFNγH, TNFHIFNγN-L, TNFN-LIFNγH, and TNFN-LIFNγN-L. Compared to patients with TNFN-LIFNγN-L, patients with TNFHIFNγH had high systemic levels of pro- and anti-inflammatory cytokines and cytotoxic molecules, and their T cells and monocytes expressed TNF receptor 1 (TNFR1). Patients with TNFHIFNγH presented the SNP rs3853839 to TLR7 and increased levels of MYD88, NFκB, and IRF7 (TLR signaling), FADD, and TRADD (TNFR1 signaling). Moreover, critical patients were observed in the four COVID-19 groups, but patients with TNFHIFNγH or TNFHIFNγN-L most required invasive mechanical ventilation. We concluded that increased TNF/IFN-γ levels are associated with hyperactive immune cells, whereas normal/low levels are associated with hypoactivity, suggesting a model to explain that the pathophysiology of critical COVID-19 may be mediated through different pathways depending on TNF and IFN-γ levels. These findings highlight the potential for exploring the modulation of TNF and IFN-γ as a therapeutic strategy in severe COVID-19.
Collapse
Affiliation(s)
- Lucero A. Ramón-Luing
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Laura Edith Martínez-Gómez
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14080, Mexico; (L.E.M.-G.); (C.M.-A.); (G.A.M.-N.); (A.L.-R.)
| | - Carlos Martinez-Armenta
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14080, Mexico; (L.E.M.-G.); (C.M.-A.); (G.A.M.-N.); (A.L.-R.)
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14080, Mexico; (L.E.M.-G.); (C.M.-A.); (G.A.M.-N.); (A.L.-R.)
| | - Karen Medina-Quero
- Immunology Laboratory, Escuela Militar de Graduados de Sanidad, Mexico City 11200, Mexico;
| | - Gloria Pérez-Rubio
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Ramcés Falfán-Valencia
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Ivette Buendia-Roldan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Julio Flores-Gonzalez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Ranferi Ocaña-Guzmán
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14080, Mexico; (L.E.M.-G.); (C.M.-A.); (G.A.M.-N.); (A.L.-R.)
| | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (L.A.R.-L.); (G.P.-R.); (R.F.-V.); (I.B.-R.); (J.F.-G.); (R.O.-G.); (M.S.)
| |
Collapse
|