1
|
Zhang Q, Zhou Q, Li H. Action and mechanisms of neferine in inflammatory diseases (Review). Mol Med Rep 2025; 32:174. [PMID: 40242976 PMCID: PMC12046375 DOI: 10.3892/mmr.2025.13539] [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: 09/28/2024] [Accepted: 02/17/2025] [Indexed: 04/18/2025] Open
Abstract
Neferine is a bisbenzylisoquinoline alkaloid derived from the seed embryo of Nelumbo nucifera, a traditional Chinese medicine. It has been extensively studied for its therapeutic potential in various disease models. Extensive research has highlighted its diverse pharmacological activities, including antitumor, anti‑inflammatory, anti‑fibrosis, anti‑oxidative stress, anti‑platelet aggregation and anti‑arrhythmic effects. The present review, however, focuses on the anti‑inflammatory properties of neferine, emphasizing its fundamental mechanisms as demonstrated in both in vivo and in vitro studies. By critically evaluating its effect on inflammation and the underlying pathways, this review aims to provide a comprehensive understanding of the potential of neferine in the management of inflammatory diseases. Furthermore, it seeks to establish a foundational framework for the future development of neferine as a novel therapeutic agent for inflammatory conditions.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Nephropathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Qiaoling Zhou
- Department of Nephropathy, Xiangya Hospital Central-South University, Changsha, Hunan 410028, P.R. China
| | - Huihui Li
- Department of Nephropathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| |
Collapse
|
2
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [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: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
3
|
Huang Y, Li G, Li D, Liu C, Chen M, Cai L, Sun M, Xu Q. Ethyl caffeate alleviates inflammatory response and promotes recovery in septic-acute lung injury via the TNF-α/NF-κB/MMP9 Axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156700. [PMID: 40220405 DOI: 10.1016/j.phymed.2025.156700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Septic acute lung injury (Septic-ALI, SA) is a severe complication of sepsis with limited clinical treatment options. Ethyl Caffeate (EC) is a phenolic compound isolated from Ilex latifolia Thunb (I. latifolia) of the Aquifoliaceae family. PURPOSE This study aimed to investigate the potential mechanisms of EC in treating SA by integrating network pharmacology and transcriptomics. METHODS We used network pharmacology to predict the potential pathways and targets of EC and validated these predictions using the GEO database, molecular docking and MDS. Subsequently, LPS-induced inflammation models in RAW cells and a mouse model of SA were established to evaluate the therapeutic effects of EC. Cell transcriptomic sequencing, along with ELISA, qRT-PCR, and Western blot analyses, were performed on both cellular and animal models to validate the key pathways and targets. RESULTS EC targeted TNF-α and MMP9, significantly alleviating LPS-induced SA through the TNF-α/NF-κB/MMP9 axis. Specifically, network pharmacology and molecular docking suggested that EC may target TNF, MMP9, EGFR, PRKACA, and MAPK3. Transcriptomic analyses, MDS and in vitro and in vivo experiments showed that EC primarily reduced the expression of p-p65 and p-IκBα in the TNF pathway by inhibiting TNF-α, thereby downregulating the expression of downstream effector molecules MMP9 and MMP14, and improving lung tissue damage, cell apoptosis, and inflammation levels in mice. CONCLUSION This study was the first to integrate network pharmacology and transcriptomic results, revealing the mechanism by which EC ameliorated SA through the TNF-α/NF-κB/MMP9 axis. Furthermore, experimental validation identified TNF-α and MMP9 as two core targets of EC, providing a valuable reference for the clinical treatment of SA.
Collapse
Affiliation(s)
- Yuanlan Huang
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Gang Li
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China.
| | - Dan Li
- Special Food Equipment Research Laboratory, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Chang Liu
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Mengying Chen
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Linli Cai
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai 200050, PR China
| | - Mingxue Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, PR China.
| | - Qingqiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, PR China; Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai 200433, PR China.
| |
Collapse
|
4
|
Lippincott MJ, Tomkinson J, Bunten D, Mohammadi M, Kastl J, Knop J, Schwandner R, Huang J, Ongo G, Robichaud N, Dagher M, Mansilla-Soto A, Saravia-Estrada C, Tsuboi M, Basualto-Alarcón C, Way GP. A morphology and secretome map of pyroptosis. Mol Biol Cell 2025; 36:ar63. [PMID: 40202832 DOI: 10.1091/mbc.e25-03-0119] [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: 04/11/2025] Open
Abstract
Pyroptosis represents one type of programmed cell death. It is a form of inflammatory cell death that is canonically defined by caspase-1 cleavage and Gasdermin-mediated membrane pore formation. Caspase-1 initiates the inflammatory response (through IL-1β processing), and the N-terminal cleaved fragment of Gasdermin D polymerizes at the cell periphery forming pores to secrete proinflammatory markers. Cell morphology also changes in pyroptosis, with nuclear condensation and membrane rupture. However, recent research challenges canon, revealing a more complex secretome and morphological response in pyroptosis, including overlapping molecular characterization with other forms of cell death, such as apoptosis. Here, we take a multimodal, systems biology approach to characterize pyroptosis. We treated human peripheral blood mononuclear cells (PBMCs) with 36 different combinations of stimuli to induce pyroptosis or apoptosis. We applied both secretome profiling (nELISA) and high-content fluorescence microscopy (Cell Painting). To differentiate apoptotic, pyroptotic, and control cells, we used canonical secretome markers and modified our Cell Painting assay to mark the N-terminus of Gasdermin D. We trained hundreds of machine learning (ML) models to reveal intricate morphology signatures of pyroptosis that implicate changes across many different organelles and predict levels of many proinflammatory markers. Overall, our analysis provides a detailed map of pyroptosis which includes overlapping and distinct connections with apoptosis revealed through a mechanistic link between cell morphology and cell secretome.
Collapse
Affiliation(s)
- Michael J Lippincott
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Jenna Tomkinson
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Dave Bunten
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | | | | | | | | | | | - Grant Ongo
- Nomic Bio, Montreal, Québec, Canada H2T 1C1
| | | | | | | | | | - Masafumi Tsuboi
- Department of Chemistry and Biotechnology, University of Tokyo, Tokyo, Japan 113-0033
| | - Carla Basualto-Alarcón
- Health Sciences Department, University of Aysén, Coyhaique, Chile
- Anatomy and Legal Medicine Department, University of Chile, Santiago, Chile
| | - Gregory P Way
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
5
|
Mahadev Bhat S, Creighton CC, Sieck GC. TNFα-mediated subcellular heterogeneity of succinate dehydrogenase activity in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2025; 328:L792-L808. [PMID: 40266735 DOI: 10.1152/ajplung.00396.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/13/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025] Open
Abstract
Tumor necrosis factor-α (TNFα) is a pro-inflammatory cytokine, which mediates acute inflammatory effects in response to allergens, pollutants, and respiratory infections. Previously, we reported that TNFα increased maximum O2 consumption rate (OCR) and mitochondrial volume density (MVD) in human airway smooth muscle (hASM) cells. However, TNFα decreased maximum OCR when normalized to mitochondrial volume. In addition, TNFα altered mitochondrial distribution and motility within hASM cells. Although high-resolution respirometry is valuable for assessing mitochondrial function, it overlooks mitochondrial structural and functional heterogeneity within cells. Therefore, a direct measurement of cellular mitochondrial function provides valuable information. Previously, we developed a confocal-based quantitative histochemical technique to determine the maximum velocity of the succinate dehydrogenase (SDH) reaction (SDHmax) in single cells and observed that cellular SDHmax corresponds with MVD. Therefore, we hypothesized that TNFα decreases SDHmax per mitochondrion in hASM cells. The hASM cells were treated with TNFα (20 ng/mL, 6 h, and 24 h) or untreated (time-matched control). Using three-dimensional (3-D) confocal imaging of labeled mitochondria and a concentric shell method for analysis, we quantified MVD, mitochondrial complexity index (MCI) and SDHmax relative to the nuclear membrane. Within each shell, SDHmax and MVD peaked in the perinuclear compartments and decreased toward the distal compartments of the cell. When normalized to mitochondrial volume, SDHmax decreased in the perinuclear compartments compared with distal compartments. TNFα caused a significant shift in mitochondrial morphometry and function compared to control. In conclusion, mitochondria within individual cells exhibit distinct morphological and functional heterogeneity, which is disrupted during acute inflammation.NEW & NOTEWORTHY Mitochondria show context-specific heterogeneity in their morphometry. Previously, we reported that acute TNFα exposure increased O2 consumption rate (OCR) and mitochondrial volume density, but decreased OCR per mitochondrion. TNFα also altered mitochondrial distribution and motility. To assess TNFα-mediated subcellular mitochondrial structural and functional heterogeneity, we used a confocal-based quantitative histochemical technique to determine the maximum velocity of succinate dehydrogenase reaction. Our findings highlight that mitochondria within cells exhibit functional heterogeneity, which is disrupted during inflammation.
Collapse
Affiliation(s)
- Sanjana Mahadev Bhat
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | | | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
6
|
Matos RCD, Bitencourt AFA, Santana PAL, de Oliveira ADM, Inoue TT, Machado BC, Caldeira ASP, Cunha Júnior ADS, Moreira CPDS, Scopel M, Machado RR. Phytochemical profile and biological activities of the hydroethanolic extract of Pouteria guianensis: A pharmacological investigation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119818. [PMID: 40245965 DOI: 10.1016/j.jep.2025.119818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The investigation of Brazil's vast plant biodiversity is crucial. The genus Pouteria, though minimally explored chemically and pharmacologically, has numerous ethnopharmacological reports. AIM OF THE STUDY This study aims to evaluate the antioxidant, anti-inflammatory, antinociceptive, angiogenic, and cytotoxic properties of hydroethanolic extract from the leaves of P. guianensis, as well as to characterize its chemical composition. MATERIAL AND METHODS Several extracts from P. guianensis leaves were evaluated for antioxidant activity using the 2,2-diphenyl-1-picrylhydrazyl (DPPH) reduction model. Chromatographic analyses and chemometric correlations were also applied. The 80 % hydroethanolic extract was selected and dereplicated using spectrometric techniques, and its anti-inflammatory and antinociceptive activities were assessed through carrageenan-induced paw edema, pain models, and the hot plate test. Antiangiogenic properties were evaluated using the chorioallantoic membrane (CAM) technique and cytokines levels were also measured. RESULTS Chemometric analyses identified myricitrin as a potential active marker for antioxidant activity, with mass spectrometry revealing a high phenolic content, particularly flavonoids quantified at 2.47 % and tannins at 17.19 %. Previous oral administration (p.o.) of the 80 % hydroethanolic extract of P. guianensis demonstrated significant anti-inflammatory and antinociceptive effects without impairing the animals' motor function. The hydroethanolic extract reduced TNF-α and IL-1β levels and also exhibited antiangiogenic properties, suggesting potential mechanisms for its anti-inflammatory action. Furthermore, no toxicity was observed in the CAM model. CONCLUSION This study highlights the significant pharmacological activities of P. guianensis hydroethanolic extract, warranting further investigation into its mechanisms of action. Additionally, it provides the first chemical characterization of the species, revealing a high phenolic content.
Collapse
Affiliation(s)
- Rafael Christian de Matos
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil; Centro Especializado Em Plantas Aromáticas, Medicinais e Tóxicas - CEPLAMT-Museu de História Natural e Jardim Botânico da Universidade Federal de Minas Gerais, Rua Gustavo da Silveira 1035, Horto, 31.080-010, Belo Horizonte, MG, Brazil.
| | - Ana Flávia Alvarenga Bitencourt
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Pedro Augusto Lemos Santana
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | | | - Thomas Toshio Inoue
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Bárbara Charine Machado
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Alisson Samuel Portes Caldeira
- Grupo Química de Produtos Naturais Bioativos, Instituto René Rachou, Fundacão, Oswaldo Cruz, Avenida Augusto de Lima 1715, Barro Preto, 30190-002, Belo Horizonte, MG, Brazil.
| | - Armando Da Silva Cunha Júnior
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Carolina Paula de Souza Moreira
- Serviço de Desenvolvimento Tecnológico Farmacêutico, Fundação Ezequiel Dias, Rua Conde Pereira Carneiro 80, Gameleira, 30510-010, Belo Horizonte, MG, Brazil.
| | - Marina Scopel
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil; Centro Especializado Em Plantas Aromáticas, Medicinais e Tóxicas - CEPLAMT-Museu de História Natural e Jardim Botânico da Universidade Federal de Minas Gerais, Rua Gustavo da Silveira 1035, Horto, 31.080-010, Belo Horizonte, MG, Brazil.
| | - Renes Resende Machado
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
7
|
Yang C, Yu W, Dang C, Zhang J, Lu J, Xue J. FEM1B enhances TRAIL-induced apoptosis in T lymphocytes and monocytes. FEBS Open Bio 2025. [PMID: 40392678 DOI: 10.1002/2211-5463.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/07/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
FEM1B is recognized for its significant pro-apoptotic function in colorectal cancer; however, its influence and mechanisms regarding apoptosis in immune cells remain inadequately elucidated. In this study, we demonstrated that FEM1B enhances TRAIL-induced apoptosis in Molt-4, Jurkat, THP-1, and U937 cell lines. Notably, the knockdown of FEM1B in transfected cells resulted in a reversal of the observed increase in cell apoptosis. Our findings indicate that FEM1B activates caspase-3 and caspase-8, but not caspase-9, in response to TRAIL stimulation, suggesting its involvement in the extrinsic caspase-dependent apoptotic pathway. Furthermore, we found that FEM1B interacted with TRAF2 and downregulates its expression in Molt-4 and Jurkat cells, thereby diminishing TRAF2's inhibitory effect on caspase-8. In THP-1 and U937 cells, FEM1B was found to upregulate TRAIL-R2, thereby promoting TRAIL-induced apoptosis. Knockout studies in murine models further corroborated that FEM1B facilitates TRAIL-induced apoptosis. These results demonstrate that FEM1B enhances TRAIL-induced apoptosis in T lymphocytes and monocytes through a caspase-dependent mechanism involving TRAF2 or TRAIL receptors.
Collapse
Affiliation(s)
- Chenbo Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenhui Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cui Dang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiahan Lu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xue
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Luo Y, Liu C, Liu J, Wang H, Fu Y, Liu Y. Exploration of Liuwei Dihuang Pill on periodontitis based on network pharmacology and molecular docking. Medicine (Baltimore) 2025; 104:e42466. [PMID: 40388786 DOI: 10.1097/md.0000000000042466] [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] [Indexed: 05/21/2025] Open
Abstract
This study explores the mechanism of Liuwei Dihuang Pill (LWDHP) in the treatment of periodontitis using network pharmacology and molecular docking. The active ingredients and targets of LWDHP were obtained from databases such as Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform. Databases such as GeneCards, OMIM, and DisGeNET were used to obtain the relevant targets related to periodontitis. The intersection of these 2 groups of targets was taken and imported into STRING to facilitate the acquisition of protein-protein interaction data, which was then imported into Cytoscape 3.10.2 to perform topological analysis to obtain the core targets. Gene ontology and Kyoto encyclopedia of genes and genomes bioinformatics enrichment analyses of the intersecting targets were performed using the DAVID database. Validation of molecular docking matching between key active ingredients with top 5 degree values and key targets with top 5 degree values in the treatment of periodontitis with LWDHP using AutoDockTools-1.5.6. A total of 69 active ingredients were discerned in LWDHP, implicating 198 periodontitis-relevant targets. Thirty-four core targets were obtained by protein-protein interaction network topology analysis, among which the key targets with the top 5 values of degree were tumor necrosis factor (TNF), serine/threonine protein kinase AKT1, sarcoma, epidermal growth factor receptor, and matrix metallopeptidase 9. Topological analysis revealed that the key active ingredients with the top 5 values of degree in LWDHP were Polyporenic acid C, Alisol B, Hydroxygenkwanin, Denudatin B, and Kadsurenone. The molecular docking results demonstrated that the binding energies of the above molecules with targets were all <-5 kcal/mol, indicating a good binding ability between these molecules. The gene ontology enrichment results indicated that the treatment of periodontitis by LWDHP was mainly related to the inflammatory response, positive regulation of phosphatidylinositol-3-kinase-Akt (PI3K-Akt) signal transduction and other processes. Analysis of the Kyoto encyclopedia of genes and genomes signaling pathway showed that the TNF signaling pathway, the PI3K-Akt signaling pathway, and so on are important signaling pathways. In conclusion, the mechanism of action of LWDHP in the treatment of periodontitis is characterized by multicomponents, multi-targets, and multi-pathways. TNF, serine/threonine protein kinase AKT1, sarcoma, epidermal growth factor receptor, and matrix metallopeptidase 9 are the key targets and the TNF signaling pathway, the PI3K-Akt signaling pathway are the key pathways. LWDHP treats periodontitis through actions such as anti-inflammatory and regulation of the balance between osteogenesis and bone destruction.
Collapse
Affiliation(s)
- Yu Luo
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Changyue Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Jiaming Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| | - Huabing Wang
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yanzhen Fu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ying Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
9
|
Choo MZY, Lim ET, Wong WSF, Chai CLL. Discovery of an NF-κB1 p105 Degrader for Anti-Inflammatory Therapy via Structural Optimization of the Coumarin Natural Product Minutuminolate. J Med Chem 2025. [PMID: 40378174 DOI: 10.1021/acs.jmedchem.5c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
In this study, the coumarin natural product minutuminolate (MNT) was used as a starting point for the development of anti-inflammatory agents. Through structure-activity relationship studies, a lead compound MD-1 was designed and synthesized, exhibiting significantly improved anti-inflammatory activities. Mechanistic studies revealed that MD-1 is a degrader of the p105 subunit of NF-κB. Gene knockdown experiments further showed that the Cullin-ring ligase (CRL) SCFβTrCP is involved in MD-1-induced p105 degradation. This leads to suppressed NF-κB transcriptional activity, which is consistent with its potent anti-inflammatory effects. Taken together, our work challenges the longstanding notion that NF-κB is undruggable, as we demonstrate that the p105 subunit of NF-κB is indeed tractable with small molecules. More importantly, our study highlights that natural products are valuable starting points for the discovery and development of anti-inflammatory agents with novel mechanisms of action.
Collapse
Affiliation(s)
- Malcolm Z Y Choo
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| | - En Tong Lim
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 16 Medical Drive, 117600 Singapore
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, 117600 Singapore
| | - Christina L L Chai
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, 117543 Singapore
| |
Collapse
|
10
|
Cui Y, Li Z, Lai M, Yang Y, Zhang Z, Feng Y, Yao M, Li J. Therapeutic effects of Saussurea graminea Dunn and its active compounds in Sepsis-associated liver injury: Transcriptomics, metabolomics and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025:119985. [PMID: 40383246 DOI: 10.1016/j.jep.2025.119985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/04/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saussurea graminea Dunn (SG), a traditional Chinese medicinal herb known as "Za Chi" in Tibet of China, is frequently utilized in the treatment of inflammatory diseases such as hepatitis. However, the active ingredients and mechanism of its therapeutic effect on Sepsis - associated liver injury (SALI) remain unclear. AIM OF THE STUDY To elucidate the effect of SG in combating SALI, uncover its mechanism of action, and explore possible active compounds. MATERIALS AND METHODS We established a SALI model by intraperitoneal injection of lipopolysaccharide to assess the efficacy of SG. Transcriptomics and metabolomics were employed to reveal its possible mechanism of action. Subsequently, Western blot, flow cytometry, confocal microscopy, quantitative PCR, HPLC-MS, and molecular docking were utilized to verify its mechanism and active ingredients. RESULTS SG effectively counteracts SALI by inhibiting the cytokine storm. Transcriptomics indicates that SG regulates SALI through mitochondrial/TNF and metabolic pathways. Metabolomics demonstrates that arachidonic acid metabolism is involved in the process of SG treating SALI. HPLC-MS identified the main components of SG as chlorogenic acid, syringin, scopoletin, rutin, isochlorogenic acid, and narcissin, and these six compounds were confirmed as potential active components in the RAW264.7 inflammation model. CONCLUSION SG and its active ingredients play a role in alleviating SALI by reducing the cytokine storm through mtDNA/TNF/arachidonic acid metabolism.
Collapse
Affiliation(s)
- Yushun Cui
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Zhiqiang Li
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Miao Lai
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Ying Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhengwen Zhang
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Yulin Feng
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Min Yao
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Junmao Li
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| |
Collapse
|
11
|
Jo HG, Seo J, Jang B, Kim Y, Kim H, Baek E, Park SY, Lee D. Integrating network pharmacology and experimental validation to advance psoriasis treatment: Multi-target mechanistic elucidation of medicinal herbs and natural compounds. Autoimmun Rev 2025:103836. [PMID: 40381707 DOI: 10.1016/j.autrev.2025.103836] [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: 04/02/2025] [Revised: 05/10/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Psoriasis, a chronic immune-mediated inflammatory disease (IMID), presents significant therapeutic challenges, necessitating exploration of alternative treatments like medicinal herbs (MH) and natural compounds (NC). Network pharmacology offers predictive insights, yet a systematic evaluation connecting these predictions with experimental validation outcomes specifically for MH/NC in psoriasis is lacking. This review specifically fills this gap by comprehensively integrating and analyzing studies that combine network pharmacology predictions with subsequent experimental validation. METHODS A systematic literature search identified 44 studies employing both network pharmacology and in vitro or in vivo experimental methods for MH/NC targeting psoriasis. This review provides a systematic analysis of the specific network pharmacology platforms, predicted targets/pathways, in vivo and in vitro experimental validation models, and key biomarker changes reported across these integrated studies. Methodological approaches and the consistency between predictions and empirical findings were critically evaluated. RESULTS This first comprehensive analysis reveals that network pharmacology predictions regarding MH/NC mechanisms in psoriasis are frequently corroborated by experimental data. Key signaling pathways, including the IL-17/IL-23 axis, MAPK, and NF-kappaB, emerge as consistently predicted and experimentally validated targets across diverse natural products. The review maps the specific network pharmacology tools and experimental designs utilized, establishing a methodological benchmark for the field and highlighting the successful synergy between computational prediction and empirical verification. CONCLUSION By systematically integrating and critically assessing the linkage between network pharmacology predictions and experimental validation for MH/NC in psoriasis, this review offers a unique clarification of the current, validated state-of-the-art, differentiating it from previous literature. It confirms network pharmacology's predictive power for natural products, identifies robustly validated therapeutic pathways, and provides a crucial benchmark, offering data-driven insights for future research into artificial intelligence-enhanced natural product-based therapies for psoriasis and other IMIDs.
Collapse
Affiliation(s)
- Hee-Geun Jo
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; Naturalis Inc., 6 Daewangpangyo-ro, Bundang-gu, Seongnam-si 13549, Republic of Korea.
| | - Jihye Seo
- Siho Korean Medicine Clinic, 407, Dongtansillicheon-ro, Hwaseong-si 18484, Republic of Korea
| | - Boyun Jang
- IntegroMediLab Co., Ltd., 143, Magokjungang-ro, Gangseo-gu, Seoul 07797, Republic of Korea
| | - Youngsoo Kim
- IntegroMediLab Co., Ltd., 143, Magokjungang-ro, Gangseo-gu, Seoul 07797, Republic of Korea
| | - Hyehwa Kim
- KC Korean Medicine Hospital, 12, Haeol 2-gil, Paju-si 10865, Republic of Korea
| | - Eunhye Baek
- RexSoft Inc., 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Soo-Yeon Park
- Department of Ophthalmology, Otolaryngology & Dermatology, College of Korean Medicine, Dongshin University, 185 Geonjae-ro, Naju-si 58245, Republic of Korea
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea.
| |
Collapse
|
12
|
Song L, Xu L, Zhang P, Li S, Qu Y, Zhao Y, Shi Z, Ma R, Li Y, Chen Y, Wang Y, Jiang Z, Wei G, Shen X. A Dual-Targeting T6SS DNase Drives Bacterial Antagonism and Eukaryotic Apoptosis via the cGAS-STING-TNF Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504086. [PMID: 40365777 DOI: 10.1002/advs.202504086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/27/2025] [Indexed: 05/15/2025]
Abstract
The Type VI secretion system (T6SS) is a key virulence mechanism utilized by many Gram-negative bacteria to mediate the microbial competition and host pathogenesis. Despite the identification of diverse T6SS effectors targeting eukaryotic or prokaryotic cells, the trans-kingdom T6SS effectors that simultaneously target both eukaryotic and prokaryotic cells remain rarely reported. In this study, it is demonstrated that Yersinia pseudotuberculosis (Yptb) T6SS secretes a DNase effector, TkeA, which induces apoptosis in host cells. The translocation of TkeA into host cells causes nuclear DNA damage. This, in turn, activates the DNA-sensing cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway. The activation of the cGAS-STING pathway by TkeA subsequently triggers apoptosis in host cells via extrinsic pathways, with tumor necrosis factor (TNF) signaling playing a critical role. Additionally, TkeA enhances bacterial competition by targeting rival bacteria, thereby promoting host colonization. These findings reveal that the transkingdom T6SS effector TkeA executes a "one weapon, two battlefields" strategy, acting as a trans-kingdom effector that enhances interbacterial competition while inducing apoptosis in host cells through the activation of the cGAS-STING-TNF axis. This highlights a previously unrecognized dimension of bacterial virulence strategies and expands the understanding of host-pathogen interactions involving T6SS effectors.
Collapse
Affiliation(s)
- Li Song
- Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi, 12100, P. R. China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Lei Xu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Pengfei Zhang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Shuying Li
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yichen Qu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yixin Zhao
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Zhenkun Shi
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Ruiqi Ma
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yongdong Li
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, 315010, P. R. China
| | - Yi Chen
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, 315010, P. R. China
| | - Yao Wang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, P. R. China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Gehong Wei
- Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi, 12100, P. R. China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Xihui Shen
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| |
Collapse
|
13
|
Zhou K, Peng K, Lin J, Wang J, Jiang Q, Ai M, Xu L, Ouyang P, Huang X, Chen D, Geng Y. ADP-heptose-LPS heptosyltransferase I (WaaC) as a key molecule maintains morphological structure and metabolic activity, simultaneously influences pathogenicity in Vibrio mimicus. Int J Biol Macromol 2025; 313:144203. [PMID: 40373916 DOI: 10.1016/j.ijbiomac.2025.144203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
ADP-heptose-LPS heptosyltransferase I (encoded by waaC gene) is a crucial enzyme in the lipopolysaccharide (LPS) synthesis, maintaining the stability of LPS and cell walls in numerous Gram-negative bacteria. Vibrio mimicus is an epidemic pathogen that threatens aquatic animals and human health, resulting in high morbidity and mortality after infecting the fish. Currently, the role of waaC gene in V. mimicus is still unclear. In this study, waaC gene deletion and complementation strains of V. mimicus were constructed. Our results show that ΔwaaC exhibited a rough phenotype on LB agar, with elevated exocrine protein and a disordered cell wall observed by the electron microscope. Transcriptomic analysis revealed that following the deletion of waaC gene, the expression of 224 genes was drastically upregulated, while the expression of 229 genes was significantly downregulated. These genes involve various pathways, including material transport, metabolism, and environmental adaptation. The tricarboxylic acid cycle (TCA cycle) and pyruvate metabolism are the primary pathways affected by waaC gene. Our phenotypic analysis is consistent with transcriptomic findings, indicating that the decreased pathogenicity of ΔwaaC is related to the effect of waaC gene on these genes, which negatively impacts V. mimicus growth, motility, adhesion, and biofilm formation while enhancing its self-aggregation. The virulence of the ΔwaaC was 103-fold lower than that of the wild strain. Moreover, the expression levels of inflammatory cytokine and pro-apoptotic genes in epithelioma papulosum cyprini (EPC) cells were dramatically upregulated during the ΔwaaC infection. These results provide valuable insights into revealing the pathogenic mechanism of V. mimicus, further bringing more options for the candidate deletion targets of the V. mimicus attenuated vaccine.
Collapse
Affiliation(s)
- Keyu Zhou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Kun Peng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jingyi Lin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jiao Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qibin Jiang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mingqi Ai
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Le Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiaoli Huang
- Department of Aquaculture, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Defang Chen
- Department of Aquaculture, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
14
|
Zhang Y, Wu Y, Ding B, Li Q, Chen X, Liu H, Xu M, Lan Y, Li Y. TNF-α inhibits Epstein Barr virus reactivation through the GPX4 mediated glutathione pathway. Sci Rep 2025; 15:16448. [PMID: 40355596 PMCID: PMC12069636 DOI: 10.1038/s41598-025-98679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
Epstein-Barr virus (EBV) is a carcinogenic γ-herpesvirus that remains latent in more than 95% of adults. The virus can undergo lytic activation when immune function is suppressed or when stimulated by drugs or pathogens. EBV reactivation poses a significant threat to human health and is closely associated with various cancers, such as Burkitt's lymphoma and nasopharyngeal carcinoma. Inhibiting EBV reactivation is a current clinical challenge. Tumour necrosis factor-α (TNF-α), an important cytokine, has different effects on various viruses. It also exerts varying effects on the same virus depending on the type of infected cell. This study aimed to investigate the impact of TNF-α on EBV reactivation and its underlying mechanisms. Our experimental research revealed that TNF-α significantly inhibits EBV reactivation and that this inhibitory effect is mediated primarily through its receptor TNFR1. Furthermore, TNF-α affects the expression of the GPX4 protein and regulates the potential ferroptosis state of cells. Using transmission electron microscopy and other methods, we observed typical characteristics of ferroptosis, such as changes in mitochondrial morphology and Fe2 + accumulation. Additionally, we established stable GPX4-knockdown cell lines, which demonstrated the crucial role of GPX4 in the process of TNF-α-mediated inhibition of EBV reactivation. Overall, TNF-α acts on the TNFR1 receptor, thereby affecting the GPX4 protein and the ferroptosis pathway to achieve its inhibitory effect on EBV reactivation. These findings provide new insights into the mechanisms of EBV reactivation and may offer new perspectives for the early treatment of EBV-related diseases.
Collapse
Affiliation(s)
- Youyu Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yilin Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Beining Ding
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qian Li
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xuenuo Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Huiling Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mingyan Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yinghua Lan
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yongguo Li
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
15
|
Srivastava V, O'Reilly C. Characteristics of cerebrospinal fluid in autism spectrum disorder - A systematic review. Neurosci Biobehav Rev 2025; 174:106202. [PMID: 40354953 DOI: 10.1016/j.neubiorev.2025.106202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Autism Spectrum Disorder (ASD) is a range of neurodevelopmental conditions characterized by impaired social interaction, learning, and restricted or repetitive behaviors. The underlying causes of ASD are still debated, but researchers have found many physiological traits like gut problems and impaired immune system to help understand the etiology of ASD. Cerebrospinal fluid (CSF) plays a critical role in maintaining the homeostasis of the neuronal environment and has, therefore, been analyzed in multiple conditions impacting the central nervous system. The study of CSF is crucial to understanding neurological disorders as its composition changes with the disorders, and these changes may indicate various disorder-related physiological mechanisms. For this systematic review, we searched PubMed, Scopus, and Web of Science for studies published between 1977 and 2025 and selected 49 studies after manual screening. We took stock of the evidence supporting the hypothesis that ASD alters the properties and composition of CSF. We systematically report on the different attributes of CSF in the ASD population that could be potential biomarkers and assist in understanding the origins and progression of ASD. We found that in CSF, immune markers, proteins, extra-axial CSF, folate, oxytocin, and vasopressin showed changes in ASD compared to the neurotypicals. We observed gaps in the literature due to variations in age and sample size and noted biases related to sex (i.e., samples are predominantly including male participants) and age (i.e., a handful of studies were conducted on adults). Our review highlights the need for more research on CSF in ASD to improve our understanding of this disorder and identify CSF biomarkers.
Collapse
Affiliation(s)
- Vandana Srivastava
- AI Institute, University of South Carolina, 5th floor, 1112 Greene St., Columbia, SC 29201, USA; Department of Computer Science and Engineering, University of South Carolina, 550 Assembly Street, Columbia, SC 29201, USA; Carolina Autism and Neurodevelopment Research Center, University of South Carolina, 1800 Gervais Street, Columbia, SC 29201, USA.
| | - Christian O'Reilly
- AI Institute, University of South Carolina, 5th floor, 1112 Greene St., Columbia, SC 29201, USA; Department of Computer Science and Engineering, University of South Carolina, 550 Assembly Street, Columbia, SC 29201, USA; Carolina Autism and Neurodevelopment Research Center, University of South Carolina, 1800 Gervais Street, Columbia, SC 29201, USA; Institute for Mind and Brain, University of South Carolina, 1800 Gervais Street, Columbia, SC 29201, USA.
| |
Collapse
|
16
|
Heidari-Japelaghi R, Valizadeh M, Haddad R. Interferon gamma-induced hub genes and key pathways: A study based on biological network analysis and experimental validation. J Biotechnol 2025; 405:72-87. [PMID: 40348089 DOI: 10.1016/j.jbiotec.2025.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
By performing a biological network analysis, we identified some hub genes, which were up- or down-regulated in the breast cancer (BC) cell line after treatment with IFN-γ. Moreover, several pathways including cytokine-cytokine receptor interaction, TNF signaling pathway, NOD-like receptor signaling pathway, and NF-κB signaling pathway were detected that their activation leads to the antiproliferation, proapoptosis, and antiviral activities. To validate in silico results, the bioactivity of recombinant human IFN-γ (hIFN-γ) produced in different hosts was analyzed by antiviral and anticancer assays. The antiviral role of the hIFN-γ preparations was evaluated by inhibition of Vesicular Stomatitis Virus (VSV)-mediated cytopathic effects on Vero cells. A dose-dependent increase in cell viability was observed at different concentrations of recombinant proteins. The maximum amount of the cell viability detected for the hIFN-γ preparations was determined at a concentration of 32.00 pg/mL. To analyze the cytotoxic efficacy of the hIFN-γ preparations on the growth and development of tumor cells, a BC cell line (MCF-7) was treated with both recombinant protein forms in a time- and dose-dependent way. The highest level of inhibiting cell proliferation was detected at a concentration of 32.00 pg/mL hIFN-γ after 72 h incubation. Anticancer and antiviral functions of IFN-γ were confirmed via the expression analysis of hub genes cd74, cxcl10, il6, and stat1 using RT-PCR. Furthermore, the hIFN-γ preparations were significantly able to up-regulate the expression of proapoptotic Bax and p53 and to down-regulate Bcl-2 as an antiapoptotic gene, showing the cytotoxic effect of hIFN-γ toward MCF-7 cells via apoptosis induction.
Collapse
Affiliation(s)
- Reza Heidari-Japelaghi
- Department of Biotechnology Engineering, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran.
| | - Mostafa Valizadeh
- Department of Plant Breeding and Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Raheem Haddad
- Department of Biotechnology Engineering, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
17
|
Huse M. Mechanoregulation of lymphocyte cytotoxicity. Nat Rev Immunol 2025:10.1038/s41577-025-01173-2. [PMID: 40312550 DOI: 10.1038/s41577-025-01173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Cytotoxic lymphocytes counter intracellular pathogens and cancer by recognizing and destroying infected or transformed target cells. The basis for their function is the cytolytic immune synapse, a structurally stereotyped cell-cell interface through which lymphocytes deliver toxic proteins to target cells. The immune synapse is a highly dynamic contact capable of exerting nanonewton-scale forces against the target cell. In recent years, it has become clear that the interplay between these forces and the biophysical properties of the target influences the entirety of the cytotoxic response, from the initial activation of cytotoxic lymphocytes to the release of dying target cells. As a result, cellular cytotoxicity has become an exemplar of the ways in which biomechanics can regulate immune cell activation and effector function. This Review covers recent progress in this area, which has prompted a reconsideration of target cell killing from a more mechanobiological perspective.
Collapse
Affiliation(s)
- Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
18
|
Cai Z, Meng K, Yu T, Xi Y, Yuan Z, Wang X, Wang C, Li L, Fu X. IFN-γ-mediated suppression of ANGPT2-Tie2 in endothelial cells facilitates tumor vascular normalization during immunotherapy. Front Immunol 2025; 16:1551322. [PMID: 40370455 PMCID: PMC12075545 DOI: 10.3389/fimmu.2025.1551322] [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: 12/25/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Tumor angiogenesis is a critical biological hallmark of cancer, which involves multiple molecularly regulated signaling pathways, including the angiopoietin (ANGPT)-Tie2 and the vascular endothelial growth factor (VEGF) signaling pathways. Despite initial optimism, targeting tumor angiogenesis in the treatment of lung adenocarcinoma (LUAD) has been unsatisfactory. Currently, monotherapy with PD-1/PD-L1 inhibitors, or their combination with bevacizumab, is considered the standard therapeutic approach for LUAD. Recent studies have shown that immunotherapy suppresses tumor angiogenesis and facilitates vascular normalization. However, whether and how anti-PD-L1 therapy influences tumor vasculature remains unclear. Methods To investigate the impact of immunotherapy on the vasculature of LUAD, a mouse model of lung adenocarcinoma was established by subcutaneous implantation of Lewis lung carcinoma cells in vivo. The effects of different treatments on microvessel density and pericyte coverage were explored, and the expression of angiogenesis-related factors was analyzed. Furthermore, to explore the molecular mechanisms through which IFN-γ regulates tumor blood vessels during immunotherapy, we elucidated the specific mechanisms in vitro by means of techniques such as siRNA, ChIP, RT-qPCR, Western blot, and immunofluorescence. Finally, the effects of IFN-γ on the proliferation, migration, and angiogenic function of endothelial cells (ECs) were evaluated through CCK-8, Transwell, and HUVEC tube formation assays. Results Employing a mouse model of LUAD, we demonstrated that PD-L1 blockade therapy inhibits tumor angiogenesis and normalizes vasculature in an IFN-γ-signaling-dependent manner. Notably, anti-PD-L1 therapy reduced Tie2 and ANGPT2 expression, and these effects were reversed by the JAK1/2 inhibitor. Mechanistically, we demonstrated that IFN-γ inhibited Tie2 and ANGPT2 expression in ECs, and suppressed ANGPT2 gene transcription through the AKT-FOXO1 signaling pathway. Interestingly, IFN-γ-mediated activation of STAT1 exerts negative regulation by directly binding to the promoter regions of the ANGPT2 and TEK genes. Functionally, IFN-γ limits the migration, proliferation, and tube formation of ECs. Discussion In conclusion, our results revealed a novel mechanism wherein IFN-γ-mediated inhibition of ANGPT2-Tie2 facilitates vascular normalization during immunotherapy in LUAD, which performs an essential function in the antitumor efficacy of immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lequn Li
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Xiangning Fu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| |
Collapse
|
19
|
Huysentruyt J, Steels W, Ruiz Pérez M, Verstraeten B, Divert T, Flies K, Lemeire K, Takahashi N, De Bruyn E, Joossens M, Brown AS, Lambrecht BN, Declercq W, Vanden Berghe T, Maelfait J, Vandenabeele P, Tougaard P. RIPK1 ablation in T cells results in spontaneous enteropathy and TNF-driven villus atrophy. EMBO Rep 2025:10.1038/s44319-025-00441-5. [PMID: 40307618 DOI: 10.1038/s44319-025-00441-5] [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: 06/09/2023] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
RIPK1 is a crucial regulator of cell survival, inflammation and cell death. Human RIPK1 deficiency leads to early-onset intestinal inflammation and peripheral T cell imbalance, though its role in αβT cell-mediated intestinal homeostasis remains unclear. In this study, we demonstrate that mice with RIPK1 ablation in conventional αβT cells (Ripk1ΔCD4) developed a severe small intestinal pathology characterized by small intestinal elongation, crypt hyperplasia, and duodenum-specific villus atrophy. Using mixed bone marrow chimeras reveals a survival disadvantage of αβT cells compared to γδT cells in the small intestine. Broad-spectrum antibiotic treatment ameliorates crypt hyperplasia and prevents intestinal elongation, though villus atrophy persists. Conversely, crossing Ripk1ΔCD4 with TNF receptor 1 Tnfr1-/- knockout mice rescues villus atrophy but not intestinal elongation. Finally, combined ablation of Ripk1∆CD4 and Casp8∆CD4 fully rescues intestinal pathology, revealing that αβT cell apoptosis in Ripk1∆CD4 drives the enteropathy. These findings demonstrate that RIPK1-mediated survival of αβT cells is essential for proximal small intestinal homeostasis. In Ripk1∆CD4 mice, the imbalanced T cell compartment drives microbiome-mediated intestinal elongation and TNF-driven villus atrophy.
Collapse
Affiliation(s)
- Jelle Huysentruyt
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wolf Steels
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mario Ruiz Pérez
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bruno Verstraeten
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tatyana Divert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kayleigh Flies
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kelly Lemeire
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nozomi Takahashi
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elke De Bruyn
- Laboratory of Microbiology, Department of Biochemistry and Microbiology, Faculty of Science, Ghent University, Ghent, Belgium
| | - Marie Joossens
- Laboratory of Microbiology, Department of Biochemistry and Microbiology, Faculty of Science, Ghent University, Ghent, Belgium
| | - Andrew S Brown
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Wim Declercq
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jonathan Maelfait
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Peter Tougaard
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
20
|
Yu X, Zhong J, Zhuang X, Lin Z, Fu H, Zhang Y. Drug-associated infections and infestations in older adults with tumor necrosis factor-alpha inhibitors: a real-world retrospective and pharmacovigilance study. Front Pharmacol 2025; 16:1533902. [PMID: 40371340 PMCID: PMC12075941 DOI: 10.3389/fphar.2025.1533902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
Objective One of the adverse events of greatest concern in patients receiving biologic therapies is the risk of infection, as infections are among the primary causes of premature mortality in this population, especially in the elderly. Because of the absence of head-to-head studies and limited duration and sample size of randomized controlled trials in the older adults, we analyzed the risk of infection associated with tumor necrosis factor (TNF) inhibitors by real-world adverse event analysis, seeking to identify medications with a reduced risk of infection and offering medication options for sensitive patients. Methods A retrospective pharmacovigilance investigation was undertaken utilizing the FDA Adverse Event Reporting System (FAERS) database from the first quarter of 2010 to the fourth quarter of 2023. Drug-associated infections and infestations associated with TNF-α inhibitors (adalimumab, infliximab, etanercept, golimumab, and certolizumab pegol) were evaluated using a disproportionality analysis. The Reporting Odds Ratio (ROR) and Bayesian Confidence Propagation Neural Network (BCPNN) were utilized to detect AE signals. Results A total case of 3,239,508 cases were included after removing duplicates. Among the protective signals, etanercept showed the lowest IC025 value in septic shock (IC025 = -3.23). Notably, golimumab showed the highest IC025 value in tuberculosis (IC025 = 2.44). The five TNF-α inhibitors have high signals in mycobacterial infectious disorders. In the stratification analysis, golimumab was associated with a highest risk of infections and infestations in ankylosing spondylitis patients (ROR = 3.07, 95%CI = 2.70-3.50; IC = 1.26, 95%CI = -0.42-2.92). Univariate and multivariate logistic regression analysis indicated that gender, weight and medicine may be influencing factors for the AEs of infections and infestations (p < 0.05). Conclusion The research highlighted that the difference in the risk of infection in the elderly who used TNF-α inhibitors between various TNF-α inhibitors, adverse events and therapeutic indications, respectively. The use of TNF-α inhibitors increased the infection risk in older adults. Etanercept exhibited the lowest infection risk, whereas certolizumab pegol manifested the highest risk in infections and infestations. Doctors need to pay close attention to the appearance of mycobacterial infectious disorders in older adults treating with TNF-α inhibitors, which displayed the strongest signal.
Collapse
Affiliation(s)
- Xihui Yu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiahong Zhong
- Department of Clinical Pharmacy, Meizhou People’s Hospital (Huangtang Hospital), Meizhou, Guangdong, China
| | - Xuemei Zhuang
- Department of Pharmacy, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Zhuomiao Lin
- Department of Clinical Pharmacy, Meizhou People’s Hospital (Huangtang Hospital), Meizhou, Guangdong, China
| | - Hongbo Fu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yaofeng Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
21
|
Dong J, Zhang J, Yao K, Xu X, Zhou Y, Zhang L, Qin C. Exploring necroptosis: mechanistic analysis and antitumor potential of nanomaterials. Cell Death Discov 2025; 11:211. [PMID: 40301325 PMCID: PMC12041361 DOI: 10.1038/s41420-025-02423-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 05/01/2025] Open
Abstract
Necroptosis, a non-apoptotic mode of programmed cell death, is characterized by the disintegration of the plasma membrane, ultimately leading to cell perforation and rupture. Recent studies have disclosed the mechanism of necroptosis and its intimate link with nanomaterials. Nanomedicine represents a novel approach in the development of therapeutic agents utilizing nanomaterials to treat a range of cancers with high efficacy. This article provides an overview of the primary mechanism behind necroptosis, the current research progress in nanomaterials, their potential use in various diseases-notably cancer, safety precautions, and prospects. The goal is to aid in the development of nanomaterials for cancer treatment.
Collapse
Affiliation(s)
- Jiaheng Dong
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
- School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Jiale Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Kunhou Yao
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, 475004, China
| | - Xiao Xu
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Yaqi Zhou
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Lei Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, 475004, China.
| | - Changjiang Qin
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, 475004, China.
| |
Collapse
|
22
|
Rahman MM, Talukder A, Rahi MS, Das PK, Grice ID, Ulett GC, Wei MQ. Evaluation of Immunostimulatory Effects of Bacterial Lysate Proteins on THP-1 Macrophages: Pro-inflammatory Cytokine Response and Proteomic Profiling. J Immunol Res 2025; 2025:2289241. [PMID: 40322557 PMCID: PMC12048194 DOI: 10.1155/jimr/2289241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Bacterial lysate proteins (BLPs) serve as potential immunostimulants, recognized by pattern recognition receptors (PRRs) on immune cells, eliciting a robust immune response. In this study, THP-1 macrophages were treated with varying doses of BLPs derived from Streptococcus pyogenes (SP), Streptococcus agalactiae (SA), and Serratia marcescens (SM). The results showed significant increases (p < 0.05) in pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-12, granulocyte macrophage-colony stimulating factor (GM-CSF), eotaxin, and macrophage inflammatory protein (MIP)-1α, except for 5 µg of all BLPs for TNF-α and eotaxin, and 5 µg of SP for IL-12 production. No significant differences were found between the corresponding doses of SP and SA or SP and SM, except for GM-CSF in all doses, while SA and SM only showed a difference at the 5 µg dose for GM-CSF. Furthermore, there were no significant differences between the 10 and 20 µg doses of all BLPs, indicating that doses higher than 10 µg do not significantly enhance the pro-inflammatory response. Combination doses of SP + SM and SA + SM did not show significant differences, except for IL-1β, suggesting no synergistic effect. Cytotoxicity was observed to increase with higher BLP concentrations in a dose-dependent manner, with combinations of SP + SM and SA + SM exhibiting greater cytotoxicity than the individual BLPs. Proteomic analysis of BLPs identified immunostimulatory proteins, including heat shock proteins (HSPs; ClpB, DnaK, and GroEL), metabolic enzymes (glyceraldehyde 3-phosphate dehydrogenase (GAPDH), enolase, and arginine deiminase (ADI)), and surface and secreted proteins (ESAT-6-like protein, CRISPR-associated endonuclease Cas9, OmpA, porin OmpC, and serralysin), which are involved in immune modulation, bacterial clearance, and immune evasion. This study underscores the potential of bacterial proteins as vaccine adjuvants or supplementary therapies; however, further research is essential to find a balance between immune activation and inflammation reduction to develop safer and more effective immunostimulants.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Chittagong, Bangladesh
| | - Asma Talukder
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Md. Sifat Rahi
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Plabon Kumar Das
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - I. Darren Grice
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C. Ulett
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ming Q. Wei
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
23
|
Shen Y, Lin P. The Role of Cytokines in Postherpetic Neuralgia. J Integr Neurosci 2025; 24:25829. [PMID: 40302252 DOI: 10.31083/jin25829] [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: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 05/02/2025] Open
Abstract
Nerve injury is a significant cause of postherpetic neuralgia (PHN). It is marked by upregulated expression of cytokines secreted by immune cells such as tumor necrosis factor alpha, interleukin 1 beta (IL-1β), IL-6, IL-18, and IL-10. In neuropathic pain (NP) due to nerve injury, cytokines are important for the induction of neuroinflammation, activation of glial cells, and expression of cation channels. The release of chemokines due to nerve injury promotes immune cell infiltration, recruiting inflammatory cytokines and further amplifying the inflammatory response. The resulting disequilibrium in neuroimmune response and neuroinflammation leads to a reduction of nerve fibers, altered nerve excitability, and neuralgia. PHN is a typical NP and cytokines may induce PHN by promoting central and peripheral sensitization. Currently, treating PHN is challenging and research on the role of cytokine signaling pathways in PHN is lacking. This review summarizes the potential mechanisms of cytokine-mediated PHN and discusses the cytokine signaling pathways associated with the central and peripheral sensitization of PHN. By elucidating the mechanisms of cytokines, the cells and molecules that regulate cytokines, and their signaling systems in PHN, this review reveals important research developments regarding cytokines and their signaling pathways mediating PHN, highlighting new targets of action for the development of analgesic drugs.
Collapse
Affiliation(s)
- Yunyan Shen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Ping Lin
- Department of Geriatrics, Hangzhou Third People's Hospital, 310009 Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Lin ZP, Gan G, Xu X, Wen C, Ding X, Chen XY, Zhang K, Guo WY, Lin M, Wang YY, Chen X, Xie C, Wang J, Li M, Zhong CQ. Comprehensive PTM profiling with SCASP-PTM uncovers mechanisms of p62 degradation and ALDOA-mediated tumor progression. Cell Rep 2025; 44:115500. [PMID: 40186868 DOI: 10.1016/j.celrep.2025.115500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/25/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
Multiple post-translational modification (PTM) proteomics typically combines PTM enrichment with multiplex isobaric labeling and peptide fractionation. However, effective methods for sequentially enriching multiple PTMs from a single sample for data-independent acquisition mass spectrometry (DIA-MS) remain lacking. We present SDS-cyclodextrin-assisted sample preparation (SCASP)-PTM, an approach that enables desalting-free enrichment of diverse PTMs, including phosphopeptides, ubiquitinated peptides, acetylated peptides, glycopeptides, and biotinylated peptides. SCASP-PTM uses SDS for protein denaturation, which is sequestered by cyclodextrins before trypsin digestion, facilitating sequential PTM enrichment without additional purification steps. Combined with DIA-MS, SCASP-PTM quantifies the proteome, ubiquitinome, phosphoproteome, and glycoproteome in HeLa-S3 cell samples, identifying serine 28 phosphorylation as a key driver of poly(I:C)-induced p62 degradation. This method also quantifies PTMs in clinical tissue samples, revealing the critical role of ALDOA K330 ubiquitination/acetylation in tumor progression. SCASP-PTM offers a streamlined workflow for comprehensive PTM analysis in both basic research and clinical applications.
Collapse
Affiliation(s)
- Zhan-Peng Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Guohong Gan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiao Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Chengwen Wen
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xin Ding
- Department of Pathology, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, Fujian 361004, China
| | - Xiang-Yu Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kaijie Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wen-Yu Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Mingxin Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yu-Yang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xi Chen
- SpecAlly Life Technology Co., Ltd., Wuhan, Hubei 430074, China
| | - Changchuan Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jinling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, No. 12 Minyou Road, Xiashan, Zhanjiang, Guangdong 524003, China.
| | - Minjie Li
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361004, China.
| | - Chuan-Qi Zhong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
25
|
Horsophonphong S, Roytrakul S, Lertruangpanya K, Kitkumthorn N, Surarit R. Proteomic analysis of dental pulp from deciduous teeth in comparison to permanent teeth: an in-vitro study. Eur Arch Paediatr Dent 2025:10.1007/s40368-025-01043-4. [PMID: 40249556 DOI: 10.1007/s40368-025-01043-4] [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: 12/24/2024] [Accepted: 03/28/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE The aims of this study were to identify proteomic profiles of dental pulp from deciduous teeth and compare the profiles of the two dentitions. METHODS Teeth that were caries-free and had normal pulp conditions were collected from twelve healthy individuals. The obtained teeth consisted of deciduous teeth (n = 6) and permanent teeth (n = 6). Proteins were extracted from pulp tissue and then analysed using liquid chromatography-tandem mass spectrometry. MaxQuant was used to identify and quantify proteins from raw mass spectrometry data of the collected deciduous and previously analysed permanent dental pulp. Differentially expressed proteins (DEPs) between the dental pulp of the two dentitions were identified by a statistical analysis conducted using Metaboanalyst with criteria P-value < 0.05 and fold change > 2. RESULTS A total of 3,636 proteins were identified in the dental pulp of deciduous teeth. The biological process functional classifications of these proteins were primarily concerned with cellular process, biological regulation, metabolic process and response to stimulus. Dental pulp protein profiles differed significantly between deciduous and permanent teeth, with 736 proteins being differentially expressed, the majority of which were highly expressed in the pulp of deciduous teeth. Pathway analysis indicated DEPs to be involved in tumour necrosis factor (TNF) signalling, nuclear factor kappa B signalling, and odontoclast/osteoclast differentiation. CONCLUSION While the dental pulp of deciduous and permanent teeth shares some characteristics, there are also significant differences in protein expression, with the TNF signalling pathway and odontoclast/osteoclast differentiation being promoted in the dental pulp of deciduous teeth.
Collapse
Affiliation(s)
- S Horsophonphong
- Department of Pediatric Dentistry, Faculty of Dentistry, Mahidol University, 6 Yothi Road, Ratchathewi, Bangkok, Thailand.
| | - S Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - K Lertruangpanya
- School of Dentistry, Mae Fah Luang University, Chiang Rai, Thailand
| | - N Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - R Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
- Faculty of Dentistry, Siam University, Bangkok, Thailand
| |
Collapse
|
26
|
Fan Y, Sekar A, McCanne M, Yuh J, Kannambadi DD, Lekkala S, Muratoglu OK, Oral E. Immune response against antibiotic-resistant and antibiotic-sensitive staphylococcus aureus in a rat model of implant infection. Sci Rep 2025; 15:13264. [PMID: 40246912 PMCID: PMC12006483 DOI: 10.1038/s41598-025-95004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Little is known about the in-vivo dynamics of biofilms associated with medical-device infections and their interplay with systemic inflammation, local immune responses, and tissue healing processes. There may be an opportunity to tailor therapeutic strategies to target these dynamics to improve treatment outcomes. We investigated immune responses to a methicillin-susceptible (ATCC 12600) and a multi-drug resistant (L1101) S. aureus strain using a rat subcutaneous implant model, analyzing local and systemic inflammation through 19 gene expressions over 21 days. Our goals were to identify differences in the immune response due to infection and also with respect to the two strains. We observed that systemic inflammation, indicated by α-2-macroglobulin, was elevated in the initial stages (up to day 7). Local inflammatory cytokine levels (IL-6, TNF-α, IL-6, TNF-α, IL-1β, IL10, IL-17, IL12a, IL12b, IFNG) varied by strain, typically higher against the clinical strain. Infections generally hindered early macrophage (MCSF1) and T-cell (CD4, CD5, CD6, CD8A) recruitment, particularly in cases involving the clinical strain. Conversely, a better healing response was observed in the infection of the more susceptible ATCC 12600 strain (VEGF, CXCR1, CXCR2, MMP-1, MMP-3, MMP-13). These results are crucial for understanding immune responses to such infections, guiding therapeutic strategies.
Collapse
Affiliation(s)
- Yingfang Fan
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
| | - Amita Sekar
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
| | - Madeline McCanne
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Jean Yuh
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | | | - Sashank Lekkala
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Orhun K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, MA, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, 55 Fruit St. GRJ 1231, Boston, MA, 02114, USA.
| |
Collapse
|
27
|
Li Y, Tu T, Luo Y, Yao X, Yang Z, Wang Y. Expression Profiles of lncRNAs and mRNAs in the Mouse Brain Infected with Pseudorabies Virus: A Bioinformatic Analysis. Viruses 2025; 17:580. [PMID: 40285022 PMCID: PMC12031243 DOI: 10.3390/v17040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Pseudorabies virus (PRV) is a pathogen that causes severe neurological dysfunction in the host, leading to extensive neuronal damage and inflammation. Despite extensive research on the neuropathogenesis of α-herpesvirus infections, many scientific questions remain unresolved, such as the largely unknown functions of long non-coding RNAs (lncRNAs) in herpesvirus-infected nervous systems. To address these questions, we used RNA sequencing (RNA-seq) to investigate the expression profiles of lncRNAs and mRNAs in the brains of mice infected with PRV. Through bioinformatic analysis, we identified 316 differentially expressed lncRNAs and 886 differentially expressed mRNAs. We predicted the biological functions of these differentially expressed lncRNAs and mRNAs using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, and the results showed that the differentially expressed transcripts were mainly involved in the innate immune response. Finally, we validated the differential expression trends of lncRNAs and mRNAs using quantitative real-time PCR (q-PCR), which were consistent with the sequencing data. To our knowledge, this is the first report analyzing the lncRNA expression profile in the central nervous system (CNS) of mice infected with PRV. Our findings provide new insights into the roles of lncRNAs and mRNAs during PRV infection of the host CNS.
Collapse
Affiliation(s)
| | | | | | | | | | - Yin Wang
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (T.T.); (Y.L.); (X.Y.); (Z.Y.)
| |
Collapse
|
28
|
Zaongo SD, Wu W, Chen Y. Pathogenesis of HIV-associated depression: contributing factors and underlying mechanisms. Front Psychiatry 2025; 16:1557816. [PMID: 40313235 PMCID: PMC12043652 DOI: 10.3389/fpsyt.2025.1557816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Cumulative evidence indicates that compared to HIV negative individuals, people living with HIV (PLWH) have a higher likelihood of developing depression, anxiety, and cognitive disorders. Depression, which is known to be a persistent and overwhelming feeling of sadness accompanied by a loss of interest in usual activities, is one of the most common mental illnesses encountered during HIV infection. Experts believe that several factors such as neuroinflammation, life stressors, lack of sleep, poor nutritional state, opportunistic infections and comorbidities, and HIV medications are contributing factors favoring the development of depression in PLWH. However, the fundamental mechanisms which underlie the involvement of these factors in the emergence of depression in the context of HIV remain poorly explored. Past researches describing the role of one or two of the preceding factors do exist; however, very few articles tackle this important topic while considering the several different putative causative factors comprehensively in the particular context of HIV infection. Herein, we elaborate on the factors currently understood to be responsible for the development of depression, and discuss the particular fundamental mechanisms whereby each factor may result in the outcome of depression. We believe that the understanding of these factors and of their underlying mechanisms is essential for the development of future therapeutic interventions to alleviate the burden of depression commonly seen in PLWH, and therefore facilitate the development of strategies to improve their overall quality of life.
Collapse
Affiliation(s)
- Silvere D. Zaongo
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Wenlin Wu
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- College of Public Health, Chongqing Medical University, Chongqing, China
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
29
|
Shi Z, Wang W, Shokrollahi B, Wang W, Abdel-Shafy H, Deng T. Role of Quercetin in Modulating Inflammation and Epigenetic Regulation of Staphylococcus aureus-Induced Bovine Mastitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8784-8797. [PMID: 40168353 DOI: 10.1021/acs.jafc.4c10550] [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: 04/03/2025]
Abstract
Mastitis is a prevalent inflammatory disease in dairy herds and presents substantial economic and welfare challenges. Although antibiotics are the most widely used and effective treatment for mastitis, research into alternative antibiotics with plant-derived compounds has gained increasing attention due to the high side effects of antibiotics. Quercetin is known to play a crucial role in regulating inflammation, yet its role in preventing and treating mastitis requires further investigation. To fill this gap, we construct a bovine mastitis model using Staphylococcus aureus (S. aureus) as the pathogen and bovine mammary epithelial cells (BMECs) as the cell model. Based on this, our study further investigated the therapeutic potential of quercetin by using in vitro assays and murine models. Our results demonstrated that quercetin inhibited the inflammatory response and reduced morphological damage in S. aureus-induced BMECs by disrupting cell adhesion. Direct RNA sequencing revealed that multiple genes enriched in the TNF/IL-17 pathway were pivotal in the ability of quercetin to mitigate inflammation, which was influenced by N6-methyladenosine (m6A) methylation. Quercetin effectively modulated CCL5 expression, a key chemokine in inflammatory responses in S. aureus-induced BMECs, through m6A methylation mediated by YTHDF2, revealing a novel epigenetic mechanism in mastitis. RNA-seq analysis showed that quercetin significantly altered genes related to inflammation, extracellular matrix regulation, and matrix metalloproteinase activity, including MMP3, MMP1, MMP1A, and IGFBP3, indicating its impact on tissue remodeling and inflammation. Additionally, quercetin disrupted S. aureus adhesion to BMECs, inhibited biofilm formation, and reduced the severity of infection. The in vivo assay supported the notion that quercetin regulates CCL5 activity to alleviate the inflammatory response in an m6A-YTHDF2-dependent manner. This study demonstrated the dual role of quercetin in inflammation suppression and epigenetic modulation via m6A, positioning quercetin as a promising therapeutic for bovine mastitis and suggesting new treatment strategies targeting CCL5- and m6A-related pathways.
Collapse
Affiliation(s)
- Zhihai Shi
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Wenjia Wang
- Guangxi Provincial Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning 530001, China
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, Henan, China
| | - Borhan Shokrollahi
- Department of Animal Science, Sanandaj Branch, Islamic Azad University, Sanandaj 6616935391, Iran
| | - Wenqi Wang
- Feed Research Institute of Xinjiang Academy of Animal Husbandry, Urumqi 830011, China
| | - Hamdy Abdel-Shafy
- Department of Animal Production, Faculty of Agriculture, Cairo University, 12613 Giza, Egypt
| | - Tingxian Deng
- Guangxi Provincial Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning 530001, China
| |
Collapse
|
30
|
Jasim MH, Saadoon Abbood R, Sanghvi G, Roopashree R, Uthirapathy S, Kashyap A, Sabarivani A, Ray S, Mustafa YF, Yasin HA. Flavonoids in the regulation of microglial-mediated neuroinflammation; focus on fisetin, rutin, and quercetin. Exp Cell Res 2025; 447:114537. [PMID: 40147710 DOI: 10.1016/j.yexcr.2025.114537] [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: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Neuroinflammation is a critical mechanism in central nervous system (CNS) inflammatory disorders, encompassing conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), traumatic brain injury (TBI), encephalitis, spinal cord injury (SCI), and cerebral stroke. Neuroinflammation is characterized by increased blood vessel permeability, leukocyte infiltration, glial cell activation, and elevated production of inflammatory mediators, such as chemokines and cytokines. Microglia act as the resident macrophages of the central nervous system, serving as the principal defense mechanism in brain tissue. After CNS injury, microglia modify their morphology and downregulate genes that promote homeostatic functions. Despite comprehensive transcriptome analyses revealing specific gene modifications in "pathological" microglia, microglia's precise protective or harmful functions in neurological disorders remain insufficiently comprehended. Accumulating data suggests that the polarization of microglia into the M1 proinflammatory phenotype or the M2 antiinflammatory phenotype may serve as a sensible therapeutic strategy for neuroinflammation. Flavonoids, including rutin, fisetin, and quercetin, function as crucial chemical reservoirs with unique structures and diverse actions and are extensively used to modulate microglial polarization in treating neuroinflammation. This paper highlights the detrimental effects of neuroinflammation seen in neurological disorders such as stroke. Furthermore, we investigate their therapeutic benefits in alleviating neuroinflammation via the modulation of macrophage polarization.
Collapse
Affiliation(s)
- Mohannad Hamid Jasim
- Biology Department, College of Education, University of Fallujah, Fallujah, Iraq.
| | - Rosull Saadoon Abbood
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India.
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq.
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq.
| |
Collapse
|
31
|
Mai Y, Huang L, Qiao Y, Qin Y, Wang L, Liao H. The Potential of Siraitia grosvenorii to Promote Bone Regeneration via Modulating Macrophage Polarization: A Network Pharmacology and Experimental Study. Int J Mol Sci 2025; 26:3609. [PMID: 40332137 PMCID: PMC12026565 DOI: 10.3390/ijms26083609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Siraitia grosvenorii (SG), a traditional Chinese medicinal herb, possesses immunomodulatory and osteoinductive properties, yet its pharmacological mechanisms in bone defect repair remain largely unexplored. This study investigates the therapeutic potential of SG through a combination of network pharmacology and experimental approaches. Active compounds were identified using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) Platform, and protein interaction targets were predicted. Molecular docking and dynamics simulations assessed interactions between SG compounds and critical targets. In vitro, RAW 264.7 macrophages treated with SG-conditioned medium exhibited enhanced M2 polarization and reduced inflammation, promoting osteogenic differentiation of co-cultured MC3T3-E1 cells as evidenced by increased alkaline phosphatase activity. In vivo, scaffolds loaded with low-dose or high-dose SG (LSG/HSG) significantly improved bone regeneration in rat calvarial defects, with HSG achieving near-complete repair and mature trabeculae at 8 weeks, alongside decreased CD86 and TNF-α levels and increased IL-10 expression. Network pharmacology identified 33 shared targets related to bone regeneration and macrophage polarization, with kaempferol and beta-sitosterol demonstrating strong binding affinities to targets such as TNF, PTGS2, and CASP3. These findings highlight the potential of SG in enhancing bone defect repair and its implications for regenerative medicine.
Collapse
Affiliation(s)
- Yuying Mai
- College of Stomatology, Guangxi Medical University, Shuangyong Road 22, Nanning 530021, China; (Y.M.); (L.H.); (Y.Q.); (Y.Q.)
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning 530021, China
| | - Linhui Huang
- College of Stomatology, Guangxi Medical University, Shuangyong Road 22, Nanning 530021, China; (Y.M.); (L.H.); (Y.Q.); (Y.Q.)
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning 530021, China
| | - Yang Qiao
- College of Stomatology, Guangxi Medical University, Shuangyong Road 22, Nanning 530021, China; (Y.M.); (L.H.); (Y.Q.); (Y.Q.)
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning 530021, China
| | - Yuan Qin
- College of Stomatology, Guangxi Medical University, Shuangyong Road 22, Nanning 530021, China; (Y.M.); (L.H.); (Y.Q.); (Y.Q.)
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning 530021, China
| | - Lufei Wang
- College of Stomatology, Guangxi Medical University, Shuangyong Road 22, Nanning 530021, China; (Y.M.); (L.H.); (Y.Q.); (Y.Q.)
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning 530021, China
| | - Hongbing Liao
- College of Stomatology, Guangxi Medical University, Shuangyong Road 22, Nanning 530021, China; (Y.M.); (L.H.); (Y.Q.); (Y.Q.)
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning 530021, China
| |
Collapse
|
32
|
Chilukuri A, Kim M, Mitra T, Gubatan JM, Urrete J, Saxon LD, Ablack A, Mikulski Z, Dobaczewska K, Shen Z, Keir M, Yi T, Kaur P, Oliveira P, Murillo-Saich J, Chang EY, Steiner CA, Jedlicka P, Guma M, Rivera-Nieves J. A Similar Mutation in the AAUU-Rich Elements of the Mouse TNF Gene Results in a Distinct Ileocolitic Phenotype: A New Strain of TNF-Overexpressing Mice. Inflamm Bowel Dis 2025; 31:1067-1081. [PMID: 39756463 PMCID: PMC11985683 DOI: 10.1093/ibd/izae307] [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: 07/17/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Tumor necrosis factor (TNF) is a pleiotropic cytokine that plays a critical role in the pathogenesis of immune-mediated diseases including inflammatory bowel disease (IBD). The stability of its mRNA transcript, determined in part by destabilizing sequences in its AAUU repeats (ARE) gene region, is an important regulator of its tissue and systemic levels. A deletion in the ARE region of the gene resulted in IBD and arthritis in mice and pigs, supporting a critical role for the cytokine in human IBD and several human arthritides. A mutation in the same area of the mouse genome by Genentech scientists (T.Y., M.K.) resulted in a similar but not identical phenotype. METHODS Here, we compare histopathological, cellular, and molecular features of the strains and propose reasons for their distinct phenotypes. First, while homozygous TNFΔARE mice develop severe arthritis and die after weaning, homozygous Genentech TNFΔARE (ΔG/ΔG) mice have normal lifespans, and males are often fertile. RESULTS We found that while the ileitic phenotype had peaked at 12 weeks of age in all mice, colitis progressed mostly after 20 weeks of age in heterozygous mice. Their variably penetrant arthritic phenotype progressed mostly after 20 weeks, also in heterozygous mice from both strains. There was expansion of central memory T and B cells in lymphoid organs of TNF-overproducing strains and their transcriptional profile shared well-known pathogenetic pathways with human IBD. Finally, we found differences in the mutated sequences within the ARE regions of the TNF gene and in their microbiota composition and genetic background. These differences likely explain their phenotypic differences. CONCLUSIONS In summary, we describe a different strain of TNF-overproducing mice with an overlapping, yet not identical phenotype, which may have differential applications than the original strain.
Collapse
Affiliation(s)
- Amruth Chilukuri
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Margaret Kim
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Taniya Mitra
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - John M Gubatan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Josef Urrete
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Leo D Saxon
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Amber Ablack
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Zbigniew Mikulski
- Microscopy and Histology Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA, USA
| | - Katarzyna Dobaczewska
- Microscopy and Histology Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA, USA
| | - Zining Shen
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Mary Keir
- Genentech Pharmaceuticals, South San Francisco, CA, USA
| | - Tangsheng Yi
- Genentech Pharmaceuticals, South San Francisco, CA, USA
| | - Prabhdeep Kaur
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Patricia Oliveira
- Rheumatology Division, University of California San Diego, La Jolla, CA, USA
| | | | - Eric Y Chang
- Radiology Department, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Calen A Steiner
- Division of Gastroenterology, University of Colorado, Denver, CO, USA
| | - Paul Jedlicka
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Mónica Guma
- Rheumatology Division, University of California San Diego, La Jolla, CA, USA
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| |
Collapse
|
33
|
Huang X, Lu J, An Y, Xu M, Chen X, Liu C, Zhou X, Shan H, Qian Y, Zhang M. Electrospun PLGA/PCL Nanofiber Film Loaded with LPA Promotes Full-Layer Wound Healing by Regulating the Keratinocyte Pyroptosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20756-20767. [PMID: 40152284 DOI: 10.1021/acsami.4c22495] [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: 03/29/2025]
Abstract
Electrospun nanofibers have a number of qualities that make them a suitable choice for skin wound healing. Lysophosphatidic acid (LPA) stimulates the keratinocytes and fibroblasts to proliferate, differentiate, and migrate and enhances skin wound healing. Here, we developed the electrospun scaffolds contained in polycaprolactone (PCL) and polylactic-co-glycolic acid (PLGA). The scaffolds loaded with LPA nanoparticles retained a porous nanofiber structure and showed better physicochemical properties and biocompatibility. The scaffold continuously releases LPA to quickly initiate cell signaling and maintain long-term anti-inflammatory activity. In this study, we found that PP scaffold with LPA reduces the disordered collagen deposition and the thickness of the newborn epidermis, improves skin healing, and reduces scar formation. Explaining the mechanism of LPA mineralized tissue regeneration in skin wound healing, LPA inhibited the pyroptosis of keratinocyte, a cell death process that induces inflammation and scar formation by inhibiting the expression of TNF-α and β-catenin proteins. Thus, the electrospun PP scaffold with LPA can be potentially developed as a therapeutic avenue to target skin wound healing.
Collapse
Affiliation(s)
- Xinqi Huang
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Jianghuiwen Lu
- Department of Medical Aesthetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, Jiangsu, China
| | - Yumei An
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Mingyuan Xu
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xueshi Chen
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Chao Liu
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xuefeng Zhou
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210000, Jiangsu, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, Jiangsu, China
| | - Yunzhu Qian
- Department of Stomatology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215123, Jiangsu, China
| | - Mingyang Zhang
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
34
|
Liu Y, Zhang H, Chen P, Liu X. Analysis of clinical factors and inflammatory cytokines in patients with lung cancer and sarcopenia: a prospective single-center cohort study. Front Oncol 2025; 15:1564399. [PMID: 40265008 PMCID: PMC12011579 DOI: 10.3389/fonc.2025.1564399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Objective To analyze the relationship between the expression of various clinical factors, inflammatory cytokines, and sarcopenia and provide new ideas for whole-course management and curative effect prediction in patients with lung cancer and sarcopenia. Methods A total of 135 patients with lung cancer recruited in the Department of Oncology, Central Hospital of Shaoyang, from January 2022 to January 2024 were analyzed and divided into sarcopenia (75 cases) and non-sarcopenia (60 cases) groups. Various statistical analyses methods were used to analyze the correlation between 4 kinds of inflammatory cytokines and sarcopenia in patients with lung cancer. Results In this study, 55.6% (75/135) of the lung cancer patients were found to have sarcopenia, with a median age of 67.3 years. Those with sarcopenia were found to be significantly associated with increased age, long duration of cigarette inhalation, and high risk of malnutrition. The results of the regression analysis indicated that long-term cigarette inhalation (odds ratio [OR]=8.187), body mass index (BMI; OR=1.356), and Nutritional Risk Screening 2002 score (OR=0.050) were statistically significant (P<0.05). Multivariable logistic regression analysis indicated that patients in the sarcopenia group were positively correlated with interleukin (IL)-6 and tumour necrosis factor (TNF)-α (P<0.05). The progression-free and overall survival of lung cancer patients with sarcopenia who received chemotherapy were significantly increased compared to those who did not receive chemotherapy(P<0.05). Conclusions Patients with a long-term cigarette inhalation, high risk of malnutrition, and low BMI have a higher probability of sarcopenia. The increased expression levels of IL-6 are positively correlated with the occurrence of sarcopenia, as well as TNF-α. The intervention of chemotherapy affects inflammatory cytokine levels. Early chemotherapy may extend the survival time of patients with lung cancer and sarcopenia.
Collapse
Affiliation(s)
- Yalan Liu
- Department of Oncology, The Central Hospital of Shaoyang, Shaoyang, China
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Hui Zhang
- Department of Oncology, The Central Hospital of Shaoyang, Shaoyang, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xinfu Liu
- Department of Oncology, The Central Hospital of Shaoyang, Shaoyang, China
| |
Collapse
|
35
|
Zou Y, Du X, Zheng X, Mao S, Xu X, Zhang S, Chen X. Fumonisin B1 induces oxidative stress, inflammation and necroptosis in IPEC-J2 cells. Vet Res Commun 2025; 49:161. [PMID: 40198395 DOI: 10.1007/s11259-025-10728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Fumonisin B1 (FB1), an important mycotoxin, poses a significant threat to public health and livestock production due to its widespread contamination. Furthermore, the gastrointestinal tract is particularly vulnerable to FB1 exposure given its frequent contamination of staple crops such as corn. Although necroptosis has been recognized as a critical mechanism underlying intestinal damage caused by certain environmental toxins, whether FB1 specifically triggers necroptosis in intestinal epithelial cells remains to be fully elucidated. In this study, the intestinal porcine epithelial cell line-J2 (IPEC-J2) was employed as an in vitro model to study the intestinal cells injury caused by FB1 and the underlying mechanisms. By measuring IPEC-J2 cell viability, intracellular reactive oxygen species, gene levels, and protein levels, it was found that FB1 dose-dependent induced IPFC-J2 cell injury, oxidative stress, and inflammation. Meanwhile, FB1 significantly increased the expression of necroptosis-related genes and proteins in IPEC-J2 cells, indicating that FB1 induced the occurrence of necroptosis. In summary, the results demonstrated FB1 can induce oxidative stress, inflammation and necroptosis in IPEC-J2 cells.
Collapse
Affiliation(s)
- Yinuo Zou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xinyu Du
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoyan Zheng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China
| | - Sichao Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xinyi Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shuxia Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
36
|
Hu X, Li Y, Liu X. Sitagliptin phosphate ameliorates chronic inflammation in diabetes mellitus via modulating macrophage polarization. Front Endocrinol (Lausanne) 2025; 16:1544684. [PMID: 40260283 PMCID: PMC12010097 DOI: 10.3389/fendo.2025.1544684] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Aim To investigate the effect and mechanism of Sitagliptin phosphate on inflammation and macrophage polarization in a mouse model of type 2 diabetes. Methods In vitro, Raw264.7 cells were cultured with a high concentration of glucose (HG) and sitagliptin phosphate (SIG). The levels of inflammatory factors and the regulation of macrophage polarization were investigated, and the differentially expressed genes between HG and HG+SIG intervention were analyzed and enriched through transcriptomics. In vivo, C57BL/6J male mice were treated with HFD+STZ to establish a type 2 diabetes mouse model were investigated the effects of regulation of macrophage polarization in the pancreas and visceral adipose tissue. Results In vitro cell experiments and transcriptomics showed that Sitagliptin phosphate decreased the secretion of inflammatory factors IL-6 and TNF-α induced by high-glucose, and increased secretion of anti-inflammatory factor IL-10 by enhancing macrophage polarization. In vivo, the body weight and abdominal visceral fat weight, the ratio of visceral fat weight to body weight and fasting blood glucose were significantly increased in the DM group compared with the Control (P<0.05), Sitagliptin phosphate treatments reversed the changes in the DM group. Moreover, histological analysis showed that compared with the Control group, the size of visceral adipocytes, hepatocyte lipid deposition and the ratio of M1/M2 macrophage were higher in the DM group, which were reversed by Sitagliptin phosphate treatments (P<0.05), insulin treatments did not have a significant effect (P>0.05). Mechanistically, Western blot showed that compared with the normal group, HG upregulated the expression of mTORc1 protein, P-65 phosphorylation and P-65 protein expression in Raw264.7 cells (P<0.05), downregulated the expression of IKKβ (P<0.05) and PPAR-γ proteins (P<0.05), Sitagliptin phosphate and insulin treatments rescued these changes. Conclusion These results indicated that Sitagliptin phosphate reduced high glucose-induced inflammation by improving the imbalance of macrophage polarization via modulating the mTORc1/ PPAR-γ/NF-κB in vitro and in vivo.
Collapse
Affiliation(s)
| | | | - Xinyue Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
37
|
Lee JJ, Yang L, Kotzin JJ, Ahimovic D, Bale MJ, Nigrovic PA, Josefowicz SZ, Mathis D, Benoist C. Early transcriptional effects of inflammatory cytokines reveal highly redundant cytokine networks. J Exp Med 2025; 222:e20241207. [PMID: 39873673 PMCID: PMC11865922 DOI: 10.1084/jem.20241207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
Inflammatory cytokines are fundamental mediators of the organismal response to injury, infection, or other harmful stimuli. To elucidate the early and mostly direct transcriptional signatures of inflammatory cytokines, we profiled all immunologic cell types by RNAseq after systemic exposure to IL1β, IL6, and TNFα. Our results revealed a significant overlap in the responses, with broad divergence between myeloid and lymphoid cells, but with very few cell-type-specific responses. Pathway and motif analysis identified several main controllers (NF-κB, IRF8, and PU.1), but the largest portion of the response appears to be mediated by MYC, which was also implicated in the response to γc cytokines. Indeed, inflammatory and γc cytokines elicited surprisingly similar responses (∼50% overlap in NK cells). Significant overlap with interferon-induced responses was observed, paradoxically in lymphoid but not myeloid cell types. These results point to a highly redundant cytokine network, with intertwined effects between disparate cytokines and cell types.
Collapse
Affiliation(s)
- Juliana J. Lee
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Liang Yang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Jonathan J. Kotzin
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dughan Ahimovic
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Michael J. Bale
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Peter A. Nigrovic
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven Z. Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
38
|
Ficagna CA, da Silva AS, Rofino RD, Zatti E, Esposito T, Xavier ACH, Wagner R, Bissacotti BF, Seghetto RB, Ternus EM, Paiano D. Effects on Performance, Immunological Response and Short-Chain Fatty Acid Profile in Feces of Nursery Piglets Fed with Organic Acids and Yeast Wall. Animals (Basel) 2025; 15:1051. [PMID: 40218444 PMCID: PMC11988164 DOI: 10.3390/ani15071051] [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: 01/08/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
The piglet nursery phase is one of the critical moments in production, especially in the first few weeks after weaning. Growth-promoting antibiotics have always been used in this phase, but the world is banning or limiting the use of antibiotics for this purpose, which has led researchers to seek alternatives, with an emphasis on organic and natural ones. As a result, this study aimed to evaluate whether a combination of organic acids and their derivatives (ammonium formate, formic acid, ammonium propionate, and acetic acid) and yeast wall (mannan oligosaccharides and beta-glucans) in piglet feeding during the nursery phase has positive effects on the SCFA profile in feces and animal health reflected in greater weight gain. A 40-day experiment was conducted in a completely randomized design containing three treatments, each with nine replicates and three piglets per replicate: negative control (NC: without additives), and a combination of organic acids and yeast wall at doses of 1 and 2 kg/ton (AO+YW-1 and AO+YW-2, respectively). Animals received four diets: pre-starter 1 (d1-7), pre-starter 2 (d8-14), starter 1 (d15-25), and starter 2 (d26-40). The animals were weighed on the day of the diet change. Blood and feces were collected on days 14 and 40 of the experiment. Piglets from the AO+YW-1 group showed more significant weight gain than NC. Feed intake was higher in piglets from the AO+YW-1 group considering the first 25 days of the experiment compared to NC; there was no treatment effect on feed conversion. Piglets from NC had higher levels of C-reactive protein and ferritin, while AO+YW-2 had higher levels of interleukin 10 and lower levels of TNF-α. A greater quantity of SCFA was observed in the feces of piglets on d40, related to the changes in propionic, butyric, isovaleric, and valeric acids. Therefore, the combination of organic acids and yeast wall used at a dose of 1 kg/ton proved to be an additive option for the diet of piglets in the nursery phase to enhance weight gain and reduce the number of doses of injectable antibiotics.
Collapse
Affiliation(s)
- Cassio Antônio Ficagna
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, Chapecó 89815-630, Brazil; (C.A.F.); (R.D.R.); (E.Z.)
| | - Aleksandro Schafer da Silva
- Departamento de Zootecnia, Universidade do Estado de Santa Catarina, Chapecó 89815-630, Brazil; (T.E.); (D.P.)
| | - Rafael Domingos Rofino
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, Chapecó 89815-630, Brazil; (C.A.F.); (R.D.R.); (E.Z.)
| | - Emerson Zatti
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, Chapecó 89815-630, Brazil; (C.A.F.); (R.D.R.); (E.Z.)
| | - Tatiane Esposito
- Departamento de Zootecnia, Universidade do Estado de Santa Catarina, Chapecó 89815-630, Brazil; (T.E.); (D.P.)
| | - Ana Carolina H. Xavier
- Departamento de Ciências de Alimento, Universidade Federal de Santa Maria, Santa Maria 97105-900, Brazil; (A.C.H.X.); (R.W.)
| | - Roger Wagner
- Departamento de Ciências de Alimento, Universidade Federal de Santa Maria, Santa Maria 97105-900, Brazil; (A.C.H.X.); (R.W.)
| | - Bianca Fagan Bissacotti
- Departamento de Zootecnia, Universidade Federal de Santa Catarina, Florianópolis 88040-900, Brazil;
| | - Ronaldo Barbieri Seghetto
- Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria 97105-900, Brazil;
| | - Eduardo Mioto Ternus
- Programa de Pós-Graduação em Ciência Animal, Universidade do Estado de Santa Catarina, Lages 88520-000, Brazil;
| | - Diovani Paiano
- Departamento de Zootecnia, Universidade do Estado de Santa Catarina, Chapecó 89815-630, Brazil; (T.E.); (D.P.)
| |
Collapse
|
39
|
Chen Y, Zhou Y, Bai Y, Jia K, Zhang H, Chen Q, Song M, Dai Y, Shi J, Chen Z, Yan X, Shen Y. Macrophage-derived CTSS drives the age-dependent disruption of the blood-CSF barrier. Neuron 2025; 113:1082-1097.e8. [PMID: 40015275 DOI: 10.1016/j.neuron.2025.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 10/09/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025]
Abstract
The choroid plexus (CP) serves as the primary source of cerebrospinal fluid (CSF). The blood-CSF barrier, composed of tight junctions among the epithelial cells in the CP, safeguards CSF from unrestricted exposure to bloodborne factors. This barrier is thus indispensable to brain homeostasis and is associated with age-related neural disorders. Nevertheless, its aging is poorly understood. Here, we report that cathepsin S (CTSS), a protease secreted from the CP macrophages, is upregulated in aged CP due to increased cell senescence. CTSS cleaves the essential tight junction component, claudin 1 (CLDN1), and, in turn, impairs the blood-CSF barrier. Notably, inhibiting CTSS or upregulating CLDN1 in aged CP rejuvenates the blood-CSF barrier and brain functions. Our findings uncover a vital interplay between immune and barrier cells that accelerates CP and brain aging, identify CTSS as a potential target to improve brain homeostasis in aged animals, and underscore the critical role of circulating proteinases in aging.
Collapse
Affiliation(s)
- Yifan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yifei Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yaqing Bai
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaiwen Jia
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qingxia Chen
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Mengjiao Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yumin Dai
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiantao Shi
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengjun Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiumin Yan
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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 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] [Track Full Text] [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
|
46
|
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
|
47
|
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
|
48
|
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
|
49
|
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
|
50
|
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
|