1
|
Liang J, Yang F, Li Z, Li Q. Epigenetic regulation of the inflammatory response in stroke. Neural Regen Res 2025; 20:3045-3062. [PMID: 39589183 PMCID: PMC11881735 DOI: 10.4103/nrr.nrr-d-24-00672] [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: 06/18/2024] [Revised: 08/15/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024] Open
Abstract
Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood-brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, long-term immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
Collapse
Affiliation(s)
- Jingyi Liang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Qian Li
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Huo C, Liu Y, Yang W, Jin Q, Liu C, Jiang Y, Zhang J, Han Y, Wang X. Identification of a dual specificity protein phosphatase and its function in regulating innate immune signaling in Crassostrea gigas. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110360. [PMID: 40268072 DOI: 10.1016/j.fsi.2025.110360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/22/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
The Pacific oyster Crassostrea gigas is one of the most important cultured bivalves in the world with high economic value. However, the healthy cultivation of oysters has been restricted by disease problems for a long time. Explore the characteristics and functions of oyster innate immune regulators help to better understand the mechanism of oyster disease resistance. Dual-specificity protein phosphatases (DUSPs) play critical roles in regulating cellular signaling during several biological processes. In this study, we identified a novel phosphatase, CgDUSP4, and investigated its regulatory role in innate immune signaling in C. gigas. Sequence analysis revealed that CgDUSP4 belongs to the MAPK phosphatase (MKP) subfamily, with a conversed kinase interaction motif at the N-terminal and a phosphatase catalytic domain at the C-terminal of the protein. CgDUSP4 was highly expressed in hemocytes and significantly upregulated in response to different pathogen-associated molecular patterns (PAMPs) stimulation. Subcellular localization analysis revealed that the protein localized in both cytoplasm and nucleus. Knock-down of CgDUSP4 affected the expression of several pro-inflammatory cytokine. CgDUSP4 protein directly interacts with CgERK, CgJNK, and Cgp38 MAPK. Furthermore, CgDUSP4 inhibits LPS induced phosphorylation of ERK MAPK. Taken together, our study reports a novel oyster innate immune regulator that responds to PAMPs stimulation and affects the expression of downstream pro-inflammatory cytokines. Moreover, it may participate in oyster innate immune regulation by inhibiting ERK signaling pathway.
Collapse
Affiliation(s)
- Chuncao Huo
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Yaqiong Liu
- School of Fisheries, Ludong University, Yantai, 264025, China.
| | - Wenhao Yang
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Qianqian Jin
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Chen Liu
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Yulu Jiang
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Jinhai Zhang
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Yijing Han
- School of Fisheries, Ludong University, Yantai, 264025, China
| | - Xiaotong Wang
- School of Fisheries, Ludong University, Yantai, 264025, China.
| |
Collapse
|
3
|
Zhang QX, Du YX, Cao JJ, Yang YB, Wu W, Xu W, Xiao BG, Xiao W. Ginsenoside Rb3 represses CPZ-induced demyelination and neuroinflammation by inhibiting TRAF6 K63 ubiquitination. Int Immunopharmacol 2025; 158:114800. [PMID: 40344978 DOI: 10.1016/j.intimp.2025.114800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Multiple sclerosis is a chronic inflammatory and neurodegenerative disorder of the central nervous system. Despite ongoing research, effective treatments remain limited, especially during progressive phase. Saponins extracted from the stem and leaf of Panax notoginseng (PNSL) demonstrate a superior anti-inflammatory effect by inhibiting NO production in LPS-induced BV2 cells. Ginsenoside Rb3, the primary active and most abundant component in PNSL, has been demonstrated to mitigate inflammation-induced damage. However, whether Rb3 mitigates demyelination by inhibiting neuroinflammation had not been previously reported. In this study, biochemical and histological assays revealed that ginsenoside Rb3 effectively mitigated Cuprizone-induced demyelination and attenuated aberrant microglial activation and reactive astrogliosis within the demyelinated areas. Mechanistic investigations demonstrated that Rb3 suppresses glial cell activation and consequently mitigates inflammatory responses by inhibiting the secretion of TNF-α, IL-6, and IL-1β. TNF receptor-associated factor 6 (TRAF6) is activated by K63-linked polyubiquitination, which leads to downstream activation of the inhibitor of nuclear factor-κB kinase (IKK) complex and mitogen-activated protein kinases (MAPKs). Furthermore, Rb3 was found to inhibit the activation of nuclear factor-κB (NF-κB) and MAPKs, as evidenced by the dephosphorylation of NF-κB p65 and the MAPKs p38 and JNK. Further investigation revealed that Rb3 binds to TRAF6 at residues 69 and 88, thereby inhibiting its K63-linked polyubiquitination. Conversely, the TRAF6 mutation at E69Q or R88N abolished the inhibition effects of Rb3 on K63-linked ubiquitination of TRAF6 and subsequent downstream signaling activation. Meta-analysis showed that Rb3 exerts its anti-inflammatory effects primarily by inhibiting the NF-κB pathway. Collectively, it is concluded that Rb3 alleviates demyelination and inhibits inflammation through bound to TRAF6 to prevent its K63-linked ubiquitination and subsequent activation of NF-κB. In this study, we have for the first time elucidated that dual mechanism by which Rb3 inhibits both NF-κB and MAPK pathways to exert its anti-inflammatory effects. This study demonstrates that Rb3 shows promising preclinical therapeutic potential. Additionally, TRAF6 represents a potential therapeutic target for MS treatment.
Collapse
Affiliation(s)
- Qian-Xia Zhang
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd, Lianyungang, China
| | - Yu-Xin Du
- Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd, Lianyungang, China
| | - Jiao-Jiao Cao
- Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd, Lianyungang, China
| | - Ying-Bo Yang
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd, Lianyungang, China
| | - Wei Wu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd, Lianyungang, China
| | - Wei Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Bao-Guo Xiao
- Department of Neurology and National Research Center for Aging and Medicine, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Wei Xiao
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd, Lianyungang, China.
| |
Collapse
|
4
|
Li Q, Li C, Liu X, Guo Z, Li X, Zhang X. The key role of Piezo1 channels in ferroptosis after spinal cord injury and the therapeutic potential of Piezo1 inhibitors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:132-140. [PMID: 40339662 DOI: 10.1016/j.pbiomolbio.2025.05.001] [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: 02/03/2025] [Revised: 04/07/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Ferroptosis has been confirmed to be one of the key mechanisms of neuronal injury and dysfunction after spinal cord injury (SCI). Mechanical stresses such as deformation, compression, and stretching not only directly cause physical damage to spinal cord tissue at the moment of SCI, but also promote the development of ferroptosis through various pathways. However, the mechanism of ferroptosis after SCI remains unclear, which hinders the development of therapeutic methods. OBJECTIVE This article aims to review the key mechanisms by which mechanical stress affects ferroptosis after SCI, including its impact on the structure and function of the endoplasmic reticulum (ER) and mitochondria, its role in triggering inflammatory responses, and its activation of mechanosensitive channels. Special emphasis is placed on the role of Piezo1 channels, which are key factors in cell mechanosensation and ion homeostasis regulation. The review explores how Piezo1 channels are upregulated by mechanical stress after SCI and participate in the ferroptosis process by mediating ion flow and other mechanisms. CONCLUSIONS Inhibiting Piezo1 channels may be a potential therapeutic strategy for SCI. This review summarizes the therapeutic potential of Piezo1 inhibitors by sorting out existing studies, hoping to provide a theoretical basis for effective therapeutic strategies targeting ferroptosis after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinyu Liu
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Zixuan Guo
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinxin Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
5
|
Wu Y, Wang XQ, Wu JY, Chen YJ, Bai JX, Li ASM, Fan XY, Wong LY, Wang L, Fu XQ, Yu ZL. A tri-compound formula comprising Ginsenoside Rg1, tetrandrine and icariin alleviates atopic dermatitis symptoms in a mouse model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156737. [PMID: 40222169 DOI: 10.1016/j.phymed.2025.156737] [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: 05/19/2024] [Revised: 03/16/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Atopic dermatitis (AD) is characterized by both IgE- and non-IgE-mediated immune responses, as well as skin barrier dysfunction. Ginsenoside Rg1, tetrandrine, and icariin each exhibit distinct properties that may contribute to the management of AD. Ginsenoside Rg1 has demonstrated efficacy in mitigating IgE-mediated allergic rhinitis, while tetrandrine is known to suppress abnormal T-cell activation. Icariin has been shown to improve intestinal barrier function, which is crucial in conditions like AD. However, the potential effectiveness of the combined formula of these compounds, referred to as GTI, in treating AD remains unexplored. PURPOSE This study aimed to investigate the anti-AD effects and mechanisms of GTI in a mouse model. METHODS A calcipotriol (MC903)-induced AD-like dermatitis mouse model was used to evaluate the anti-AD effects of GTI. Dermatitis scores and mouse ear thickness were recorded to assess disease severity. Ear tissues, ear-draining lymph nodes, spleens and sera were collected for use in the investigation of the effects and mechanisms of action of GTI. RESULTS Topical application of GTI significantly alleviated AD-like dermatitis in mice, as evidenced by decreased dermatitis scores, reduced ear thickening, and diminished epidermal and dermal thickness, along with lower levels of the inflammatory cytokines IL-1β and IL-4 in ear tissues. Unlike the positive dexamethasone, GTI had no significant toxicity in the model mice. Topical GTI lowered serum IgE levels and diminished the accumulation of eosinophils and mast cells in ear tissues of model mice, suggesting that GTI mitigates IgE-mediated allergic reactions. GTI significantly decreased the numbers of CD4+ T cells in ear tissues, ear-draining lymph nodes and the spleen, demonstrating its suppressive effect on hyperactive immune responses. The protein levels of ZO-1 and claudin-1, two tight junction proteins, were elevated in the ear tissues of mice treated with GTI, indicating a beneficial effect of this formula on skin barrier function. Additionally, GTI inhibited the activation of mitogen-activated protein kinases (MAPKs), as indicated by the downregulation of phospho-p38 (Thr180/182), phospho-ERK (Thr202/Tyr204), and phospho-JNK (Thr183/185) protein levels in mouse ear tissues. CONCLUSION This study, for the first time, demonstrated that the topical application of GTI alleviates symptoms of AD without overt toxicity in a calcipotriol-induced AD mouse model. The anti-AD effects of GTI are associated with the suppression of allergic reactions, reduction of hyperactive immune responses, improvement of skin barrier function, and inhibition of MAPK activation. These findings suggest that GTI has the potential to be developed into a safe and effective treatment for AD.
Collapse
Affiliation(s)
- Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiao-Qi Wang
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jia-Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ying-Jie Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jing-Xuan Bai
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Amy Sze-Man Li
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiao-Yun Fan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Lut-Yi Wong
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Li Wang
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiu-Qiong Fu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Zhi-Ling Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; JaneClare Transdermal TCM Therapy Laboratory, Hong Kong Baptist University, Hong Kong SAR, China; Research and Development Centre for Natural Health Products, HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
6
|
Chen YC, Takada M, Nagornyuk A, Yu M, Yamada H, Nagashima T, Ohtsuka M, DeLuca JG, Markus SM, Takaku M, Suzuki A. Inhibition of p38-MK2 pathway enhances the efficacy of microtubule inhibitors in breast cancer cells. eLife 2025; 13:RP104859. [PMID: 40439108 PMCID: PMC12122001 DOI: 10.7554/elife.104859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025] Open
Abstract
Microtubule-targeting agents (MTAs) are widely used as first- and second-line chemotherapies for various cancers. However, current MTAs exhibit positive responses only in subsets of patients and are often accompanied by side effects due to their impact on normal cells. This underscores an urgent need to develop novel therapeutic strategies that enhance MTA efficacy while minimizing toxicity to normal tissues. Here, we demonstrate that inhibition of the p38 MAPK-MK2 signaling pathway sensitizes cancer cells to MTA treatment. We utilize CMPD1, a dual-target inhibitor, to concurrently suppress the p38-MK2 pathway and microtubule dynamicity. In addition to its established role as an MK2 inhibitor, we find that CMPD1 rapidly induces microtubule depolymerization, preferentially at the microtubule plus end, leading to the inhibition of tumor growth and cancer cell invasion in both in vitro and in vivo models. Notably, 10 nM CMPD1 is sufficient to induce irreversible mitotic defects in cancer cells, but not in non-transformed normal cells, highlighting its high specificity to cancer cells. We further validate that a specific p38-MK2 inhibitor significantly potentiates the efficacy of subclinical concentrations of MTA. In summary, our findings suggest that the p38-MK2 pathway presents a promising therapeutic target in combination with MTAs in cancer treatment.
Collapse
Affiliation(s)
- Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Mamoru Takada
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Aerica Nagornyuk
- Department of Biomedical Science, University of North Dakota School of Medicine and Health ScienceGrand ForksUnited States
| | - Muhan Yu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Hideyuki Yamada
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Takeshi Nagashima
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State UniversityFort CollinsUnited States
| | - Steven M Markus
- Department of Biochemistry and Molecular Biology, Colorado State UniversityFort CollinsUnited States
| | - Motoki Takaku
- Department of Biomedical Science, University of North Dakota School of Medicine and Health ScienceGrand ForksUnited States
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Carbone Comprehensive Cancer Center, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
7
|
Liu Y, Wu Z, Gu C, Fang J, Peng Y, Peng L, Chen W, Yao L, He L. ShenJiaoLingCao decoction ameliorates cyclophosphamide-induced splenic injury and immunosuppression via the inhibition of MEK/ERK signaling pathway activity and modulation of amino acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119830. [PMID: 40250640 DOI: 10.1016/j.jep.2025.119830] [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: 12/04/2024] [Revised: 03/20/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE ShenJiaoLingCao Decoction (SJLCD) is derived from the classic Chinese medicine prescription, which consists of ten kinds of herbs. In China, SJLCD has been used as an immunomodulator in clinical practice for more than ten years. However, no relevant studies have been done to clarify the pharmacodynamic underpinnings of its regulation of the body's immune system and its related processes. AIM OF THE STUDY This study aims to assess the immunomodulatory effects of SJLCD. MATERIALS AND METHODS Ultra performance liquid chromatography-quadrupole-orbitrap mass spectrometry (UPLC-Q-Orbitrap MS) was utilized to characterize the chemical constituents in SJLCD and establish its fingerprint profile. Predicting potential bioactive compounds in SJLCD for immunomodulatory effects and elucidating their mechanisms of action using artificial intelligence technology. Experiments at the animal level were carried out to verify the accuracy of the predictions. Firstly, an immunocompromised model was constructed by intraperitoneal injection of 80 mg/kg of cyclophosphamide (CTX) into rats for 3 consecutive days, and SJLCD was administered by oral administration for 14 days. The immunomodulatory effect of SJLCD on immune organs was verified by evaluating the immune organ index and histopathological examinations using hematoxylin and eosin (H&E) staining. The effect of SJLCD on relevant immune cells was examined by measuring erythrocytes, leukocytes and lymphocytes. The effect of SJLCD on relevant immune molecules was assessed by detecting the levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β), matrix metalloproteinase-9 (MMP9), cluster of differentiation 3 (CD3), cluster of differentiation 4 (CD4) and cluster of differentiation 8 (CD8). Western blot was used to verify and analyze the possible immunomodulatory mechanisms of SJLCD. Finally, serum untargeted metabolomics was used to detect the differential metabolites of SJLCD in immunocompromised rats. RESULTS In this study, a total of 91 compounds were identified in the SJLCD, and the results showed a high degree of similarity (S1-S11 > 0.935) among the 11 samples in positive ion mode. Artificial intelligence computer techniques predicted that quercetin, kaempferol, and fumarine in SJLCD bound better to core targets, especially MAPK1. On animal-level validation, it was found that from an immune organ perspective, SJLCD ameliorated CTX-induced thymus and spleen damage. From an immune cell perspective, SJLCD significantly increased peripheral erythrocyte, leukocyte and lymphocyte counts in immunocompromised rats. From the immune molecular level, SJLCD down-regulated the levels of TNF-α, IL-6, IL-1β, MMP9, CD8 and up-regulated the level of CD3 and CD4 which normalize its secretion. Mechanistically, SJLCD regulates immunity possibly through the MEK/ERK signaling pathway and by affecting amino acid metabolism. CONCLUSION In the present study, we found that SJLCD has satisfactory immunomodulatory activity, which may be achieved by affecting the MEK/ERK signaling pathway and amino acid metabolism of the body.
Collapse
Affiliation(s)
- Yuzhen Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China
| | - ZhuXia Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China
| | - Chen Gu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China
| | - Jing Fang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China
| | - Yusi Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China
| | - Lei Peng
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; He Rongjia Pharmaceutical Technology Co., Nantong, 226000, Jiangsu, China
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, Anhui, China
| | - Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China.
| | - Ling He
- Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, 230012, China.
| |
Collapse
|
8
|
Richter HI, Gover O, Hamburg A, Bendalak K, Ziv T, Schwartz B. Impact of Black Soldier Fly Larvae Oil on Immunometabolic Processes. Int J Mol Sci 2025; 26:4855. [PMID: 40429995 PMCID: PMC12112032 DOI: 10.3390/ijms26104855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
The oil extract derived from black soldier fly (Hermetia illucens) larvae (BSFL) is characterized by a distinctive fatty acid composition and bioactive compounds with demonstrated anti-inflammatory properties, as shown in our previous work. The present study aims to mechanistically explore the immunomodulatory effects of a saponified form of BSFL oil (MBSFL) and its potential interaction with metabolic signaling pathways. Using Pam3CSK4-polarized M1 primary human peripheral blood mononuclear cells (PBMCs), we demonstrate that MBSFL phenotypically suppressed the secretion of pro-inflammatory cytokines TNFα, IL-6, IL-17, and GM-CSF (p < 0.01) without altering anti-inflammatory cytokine levels (TGFβ1, IL-13, and IL-4). A phosphoproteomic analysis of Pam3CSK4-stimulated THP-1 macrophages revealed MBSFL-mediated downregulation of CK2 and ERK kinases (p < 0.05), key regulators of NF-κB signaling activation. We confirmed that MBSFL directly inhibits NF-κB p65 nuclear translocation (p < 0.05), using both immunofluorescence staining and a western blot analysis of nuclear and cytoplasmic fractions. In the context of metabolism, using a luciferase reporter assay, we demonstrate that MBSFL functions as a weak agonist of PPARγ and PPARδ (p < 0.05), which are nuclear receptors involved in lipid metabolism and immune regulation. However, subsequent immunoblotting revealed a macrophage polarization-dependent regulation: MBSFL upregulated PPARγ in M0 macrophages but did not prevent its suppression upon Pam3CSK4 stimulation, whereas it specifically enhanced PPARδ expression during M1 polarization (p < 0.05). This study provides novel experimental evidence supporting our hypothesis of MBSFL's role in immunometabolism. We demonstrate for the first time that MBSFL acts as a dual regulator by suppressing NF-κB-mediated inflammation while promoting PPARδ activity-an inverse relationship with potential relevance to immunometabolic disorders.
Collapse
Affiliation(s)
- Hadas Inbart Richter
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ofer Gover
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Amit Hamburg
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Keren Bendalak
- Smoler Proteomics Center, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Betty Schwartz
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
9
|
Han Y, Wang S, Xiong Y, Sha T, Xiong Z, Li S, She W, Zhang Y, He X, Zou S, Cheng J, Meng J, Yuan Q, Huang L, Xie Y, Tao L, Peng Z. Peroxiredoxin-1 aggravates hypoxia-induced renal injury by promoting inflammation through the TLR4/MAPK/NF-κB signaling pathway. Free Radic Biol Med 2025:S0891-5849(25)00682-3. [PMID: 40398686 DOI: 10.1016/j.freeradbiomed.2025.05.399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/28/2025] [Accepted: 05/19/2025] [Indexed: 05/23/2025]
Abstract
Hypoxia can induce pathological alterations to the kidneys, such as activation of inflammatory signaling pathways. This form of inflammation is pathogen-free and is referred to as aseptic inflammation. Currently, the mechanisms leading to aseptic inflammation under hypoxia are not well understood. Emerging evidence has indicated that Prdx1, a member of the peroxidase family, contributes to the development of various diseases by stimulating aseptic inflammation. This study was conducted to reveal the potential role of Prdx1 in the pathogenesis of hypoxia-induced renal injury. A mouse model of systemic hypoxia was developed, which revealed that Prdx1 levels were elevated in injured kidneys and peripheral circulation. A comparable increase was also observed in hypoxia-treated immortalized bone marrow-derived macrophages (iBMDMs). Knock-down of Prdx1 in mice caused a significant reduction in renal tissue injury and inflammation induced by hypoxic injury. In addition, we demonstrated that Prdx1 modulates inflammatory responses by activating the TLR4/MAPK/NF-κB signaling pathways. Recombinant Prdx1 promoted the activation of these pathways in macrophages, whereas genetic knockout of Prdx1 or pharmacological inhibition suppressed their activity. Altogether, we found a previously unrecognized role for Prdx1 in the regulation of inflammation in hypoxia-induced renal injury. These findings suggest that Prdx1 can be a potential target for treating this severe disease.
Collapse
Affiliation(s)
- Yuanyuan Han
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Songkai Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Yiwei Xiong
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Tu Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital,Guangzhou Guangdong,China
| | - Zujian Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha Hunan, China
| | - Shenglan Li
- Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China; Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha Hunan, China
| | - Wenzhe She
- Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China; Department of Cell biology, School of Life Sciences, Central South University, Changsha Hunan, China
| | - Yan Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Xin He
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Sijue Zou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Jiawei Cheng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Jie Meng
- Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China; Department of Respiratory Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Ling Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha Hunan, China; Key Laboratory of Organ Fibrosis of Hunan Province, Central South University, Changsha Hunan, China.
| |
Collapse
|
10
|
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; 24:103836. [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-κB, 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
|
11
|
Peng P, Sun J, Li MS, Cheng RX, Liu SQ, Qin MB, Zhang JX, Huang JA. SPDL1 inhibition enhances colorectal cancer progression via epidermal growth factor receptor/extracellular signal-regulated kinase pathways. World J Gastrointest Oncol 2025; 17:104686. [DOI: 10.4251/wjgo.v17.i5.104686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/11/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND In patients with colorectal cancer (CRC), tumour metastasis is the leading cause of death. The search for key genes involved in metastasis of CRC is imperative for improved prognoses and treatments. SPDL1 has been implicated in the development of CRC, however, its mechanism of action remains unclear.
AIM To investigate the role and mechanism of action by which SPDL1 inhibits the development and metastasis of CRC.
METHODS In this study, we examined the relationship between SPDL1 expression and CRC prognosis using immunohistochemistry. Survival analyses were performed using Kaplan-Meier analysis and log-rank test. After knocking down SPDL1 in the HCT116 cancer cell line changes in cell viability, migration, invasion, and gene expression were examined using a cell counting kit 8 assay, Transwell assay, and Western blot. The effect of SPDL1 on the cell cycle was assessed using flow cytometry. RNA sequencing was used to analyse the effect of SPDL1 on gene expression of CRC cells. The mechanism of action of SPDL1 in CRC was further clarified using U0126, an inhibitor of the mitogen-activated protein kinase signaling pathway.
RESULTS SPDL1 is expressed at low levels in tissues of patients with CRC, and this reduced expression is associated with poor prognosis. Functionally, low expression of SPDL1 in CRC promotes cell proliferation, migration, invasion, and affects the cell cycle. Mechanistically, SPDL1 affects the progression of CRC through its regulation of the process of epithelial-mesenchymal transition (EMT) and of the epidermal growth factor receptor (EGFR)/ extracellular signal-regulated kinase (ERK) signaling pathways.
CONCLUSION This study showed that the loss of SPDL1 may induce EMT and promote cell migration and invasion in CRC through the EGFR/ERK pathway.
Collapse
Affiliation(s)
- Peng Peng
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Juan Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Meng-Shi Li
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Ruo-Xi Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Shi-Quan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Meng-Bin Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Jin-Xiu Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Jie-An Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
12
|
Jeon H, Oh JY, Ahn S, Yeom M, Ha IJ, Son HS, Park SE, Park J, Huh E, Baek IY, Nam MH, Na C, Oh MS, Park HJ. Invasive laser acupuncture targeting muscle: a novel approach to protect dopaminergic neurons and reduce neuroinflammation in a brain of Parkinson's disease model. Chin Med 2025; 20:59. [PMID: 40336061 PMCID: PMC12057028 DOI: 10.1186/s13020-025-01104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Parkinson's disease (PD) affects 1-2% of the global population and presents significant therapeutic challenges. Due to the limitations of existing treatments, there is a pressing need for alternative approaches. This study investigated the effects of invasive laser acupuncture (ILA), which combines acupuncture and photobiomodulation. In this method, optical fibers are inserted into the muscle layers of the acupoint to enhance therapeutic outcomes. Mice with MPTP-induced PD were treated with ILA at 830 nm or 650 nm. Protective effects of nigrostriatal dopaminergic neurons and fibers were assessed by examining TH immunoreactivity in the brain. Neuroinflammation markers in the brain and muscle metabolomic profiles were also analyzed. Comparisons between invasive and non-invasive laser application, as well as the impact of nerve blocking with lidocaine, were also evaluated. ILA at 830 nm (ILA830) significantly improved motor performance and increased the nigrostriatal TH-positive immunoreactivities. It reduced the levels of α-synuclein, apoptotic proteins, and inflammatory cytokines, while increasing anti-inflammatory in the brain. ILA830 also decreased nigrostriatal astrocyte and microglia activation. Muscle metabolomic analysis showed distinct group clustering and significant changes in metabolites like glucose and galactose, correlating with improved motor functions. Invasive laser treatment was more effective than non-invasive, and lidocaine pre-treatment did not block its effects. ILA at 830 nm effectively ameliorates PD symptoms by protecting dopaminergic neurons, and reducing neuroinflammation in the brain. Muscle metabolomic changes by ILA830, such as increased glucose and galactose, correlate with motor improvement. This approach offers a promising strategy for PD treatment, warranting further research to optimize its use in clinical settings.
Collapse
Affiliation(s)
- Halin Jeon
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Department of KHU-KIST Convergence Science Technology, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Ju-Young Oh
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Sora Ahn
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - In Jin Ha
- Korean Medicine Clinical Trial Center, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Hong-Seok Son
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Seong-Eun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Juhan Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Eugene Huh
- Department of Formulae Pharmacology, College of Korean Medicine, Gachon University, Sujeong-gu, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - In-Yeop Baek
- Department of KHU-KIST Convergence Science Technology, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Min-Ho Nam
- Department of KHU-KIST Convergence Science Technology, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Changsu Na
- Department of Acupoint and Meridian, Korean Medical College, Dongshin University, Naju-si, Jeollanam-do, 58245, Republic of Korea
| | - Myung Sook Oh
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
- Department of KHU-KIST Convergence Science Technology, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
- Department of Anatomy and Information Science, College of Korean Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
13
|
Bai X, Guo YR, Zhao ZM, Li XY, Dai DQ, Zhang JK, Li YS, Zhang CD. Macrophage polarization in cancer and beyond: from inflammatory signaling pathways to potential therapeutic strategies. Cancer Lett 2025; 625:217772. [PMID: 40324582 DOI: 10.1016/j.canlet.2025.217772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Macrophages are innate immune cells distributed throughout the body that play vital roles in organ development, tissue homeostasis, and immune surveillance. Macrophages acquire a binary M1/M2 polarized phenotype through signaling cascades upon sensing different signaling molecules in the environment, thereby playing a core role in a series of immune tasks, rendering precise regulation essential. M1/M2 macrophage phenotypes regulate inflammatory responses, while controlled activation of inflammatory signaling pathways is involved in regulating macrophage polarization. Among the relevant signaling pathways, we focus on the six well-characterized NF-κB, MAPK, JAK-STAT, PI3K/AKT, inflammasome, and cGAS-STING inflammatory pathways, and elucidate their roles and crosstalk in macrophage polarization. Furthermore, the effects of many environmental signals that influence macrophage polarization are investigated by modulating these pathways in vivo and in vitro. We thus detail the physiological and pathophysiological status of these six inflammatory signaling pathways and involvement in regulating macrophage polarization in cancer and beyond, as well as describe potential therapeutic approaches targeting these signaling pathways. In this review, the latest research advances in inflammatory signaling pathways regulating macrophage polarization are reviewed, as targeting these inflammatory signaling pathways provides suitable strategies to intervene in macrophage polarization and various tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Xiao Bai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yun-Ran Guo
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhe-Ming Zhao
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Jia-Kui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yong-Shuang Li
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Chun-Dong Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
14
|
Wang J, Li S, Ye J, Yan Y, Liu Q, Jia Q, Jia Y, Wang L. Mesencephalic astrocyte-derived neurotrophic factor (MANF): A novel therapeutic target for chemotherapy-induced peripheral neuropathy via regulation of integrated stress response and neuroinflammation. Neuropharmacology 2025; 268:110342. [PMID: 39909174 DOI: 10.1016/j.neuropharm.2025.110342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/20/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) represents a severe complication, impacting up to 90% of cancer patients administered with chemotherapeutic agents such as oxaliplatin. The purpose of our study was to examine the potential role and therapeutic efficacy of Mesencephalic Astrocyte-derived Neurotrophic Factor (MANF), given its recognized neuroprotective and immunomodulatory properties in diverse neurological disorders. Utilizing an oxaliplatin-induced CIPN mouse model, we investigated MANF expression in the dorsal root ganglia (DRG) and spinal cord, and evaluated the impacts of AAV-mediated MANF overexpression on CIPN. Our findings revealed substantial downregulation of MANF expression in both the DRG and spinal cord of CIPN inflicted mice, with MANF majorly localized in neurons as opposed to glial cells. Intrathecal administration of AAV-MANF preceding oxaliplatin treatment yielded several beneficial results. MANF overexpression diminished mechanical hypersensitivity and decreased Calcitonin Gene-Related Peptide (CGRP) expression in DRG and the spinal dorsal horn. These enhancements were concomitant with modulation of the integrated stress response (ISR) and neuroinflammation. Intervention with AAV-MANF effectively regulated ISR markers (BiP, CHOP, and p-eIF2α), mitigated activation of microglia and astrocytes in the DRG and spinal dorsal horn, and inhibited NFκB and ERK inflammatory signaling pathways. To conclude, our study underscores the potential of MANF as a viable therapeutic target for CIPN, manifesting its ability to modulate ISR and neuroinflammation. These insights recommend that continued exploration of MANF-centered approaches could facilitate the advancement of more efficacious interventions for this incapacitating chemotherapy complication.
Collapse
Affiliation(s)
- Juan Wang
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Shenghong Li
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Jishi Ye
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yafei Yan
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Qi Liu
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Qiang Jia
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yifan Jia
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| | - Long Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
15
|
Niu M, Wang YZ, Deng XM, Wu X, Hua ZY, Lv TT. Tryptanthrin alleviate lung fibrosis via suppression of MAPK/NF-κB and TGF-β1/SMAD signaling pathways in vitro and in vivo. Toxicol Appl Pharmacol 2025; 498:117285. [PMID: 40089192 DOI: 10.1016/j.taap.2025.117285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF), a progressive interstitial lung disease of unknown etiology, remains a therapeutic challenge with limited treatment options. This study investigates the therapeutic potential and molecular mechanisms of Tryptanthrin, a bioactive indole quinazoline alkaloid derived from Isatis tinctoria L., in pulmonary fibrosis. In a bleomycin-induced murine IPF model, Tryptanthrin administration (5 and 10 mg/kg/day for 28 days) significantly improved pulmonary function parameters and attenuated histological evidence of fibrosis. Mechanistic analysis revealed dual pathway modulation: Tryptanthrin suppressed MAPK/NF-κB signaling through inhibition of phosphorylation events, subsequently reducing pulmonary levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). Concurrently, it attenuated TGF-β1/Smad pathway activation by decreasing TGF-β1 expression and Smad2/3 phosphorylation, thereby downregulating fibrotic markers including COL1A1, α-smooth muscle actin (α-SMA), and fibronectin in lung tissues. Complementary in vitro studies using Lipopolysaccharide (LPS) or TGF-β1-stimulated NIH3T3 fibroblasts confirmed these anti-inflammatory and anti-fibrotic effects through analogous pathway inhibition. Our findings demonstrate that Tryptanthrin exerts therapeutic effects against pulmonary fibrosis via coordinated modulation of both inflammatory (MAPK/NF-κB) and fibrotic (TGF-β1/Smad) signaling cascades, suggesting its potential as a novel multi-target therapeutic agent for IPF management.
Collapse
Affiliation(s)
- Min Niu
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China.
| | | | - Xiang-Min Deng
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| | - Xin Wu
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| | - Zheng-Ying Hua
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| | - Ting-Ting Lv
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| |
Collapse
|
16
|
Zhao L, Li X, Gao M, Liu L, Ma B, Liu X, Zhang J, Liu R, Du B, Wei R, Nian H. M6A Modified miR-31-5p Suppresses M1 Macrophage Polarization and Autoimmune Dry Eye by Targeting P2RX7. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415341. [PMID: 40068094 PMCID: PMC12061282 DOI: 10.1002/advs.202415341] [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] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/03/2025] [Indexed: 05/10/2025]
Abstract
The dysregulation of the M1/M2 macrophage balance plays a pivotal role in autoimmune diseases. However, the interplay between microRNAs (miRNAs) and N6-methyladenosine (m6A) modulation in regulating this balance remains poorly understood. Here, a significant reduction in miR-31-5p levels is observed in the lacrimal glands of rabbit autoimmune dacryoadenitis and the peripheral blood mononuclear cells (PBMCs) of Sjögren's syndrome (SS) dry eye patients. Overexpression of miR-31-5p exhibits preventive and therapeutic effects on rabbit autoimmune dacryoadenitis. Further investigation revealed that miR-31-5p overexpression significantly restored the M1/M2 macrophage balance both in vivo and in vitro. Mechanistically, miR-31-5p directly targets the P2x7 receptor (P2RX7), leading to the inactivation of p38 mitogen-activated protein kinases (MAPK) signaling and reduced expression of M1 markers. Furthermore, methylated RNA immunoprecipitation and luciferase reporter assays demonstrated that fat mass and obesity-associated protein (FTO)-mediated m6A demethylation, which sustains pri-miR-31 stability, is responsible for the decreased miR-31-5p levels in autoimmune dry eye. Notably, PBMC samples from SS dry eye patients further support the link between reduced miR-31-5p levels and M1 macrophage activation observed in rabbits. Overall, these data highlight the critical role of the FTO/miR-31-5p/P2RX7/p38 MAPK axis in autoimmune inflammation, suggesting their potential as therapeutic targets for autoimmune dry eye.
Collapse
Affiliation(s)
- Lu Zhao
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Xuejia Li
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Min Gao
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Lin Liu
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Binyun Ma
- Department of Medicine/HematologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
| | - Xun Liu
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Jiachen Zhang
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Ruoxuan Liu
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Bei Du
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and DiseasesTianjin Branch of National Clinical Research Center for Ocular DiseaseEye Institute and School of OptometryTianjin Medical University Eye HospitalTianjin300384China
| |
Collapse
|
17
|
He Y, Xiao D, Zhu H, Chen C, Liu Q, Xie J, Wei L, Dai Y, Ning Y, Li Y. Notch Signaling Aggravates Helicobacter pylori-Induced Inflammation by Promoting Macrophage Activation and Proinflammatory Th1/Th17 Responses. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00148-8. [PMID: 40316214 DOI: 10.1016/j.ajpath.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 05/04/2025]
Abstract
The role of Notch signaling in regulating the immune response in infectious and inflammatory diseases has been extensively reported. However, its specific involvement in Helicobacter pylori infection is yet to be fully understood. In this study, in vitro analysis utilizing real-time quantitative PCR and Western blot revealed that H. pylori triggers the activation of Notch signaling in murine bone marrow-derived macrophages (BMDMs) and co-cultured CD4+ T cells, a process mediated by the Notch ligand protein jagged-1 (Jag1). There was a reciprocal enhancement between Jag1-Notch signaling and NF-κB pathway in H. pylori-infected macrophages. Pretreatment with a Notch signaling inhibitor, DAPT, reduced the expression of inflammatory mediators in macrophages, modulated their phenotype, and inhibited Th1 differentiation. In vivo, after treatment with DAPT in H. pylori-infected mice, the differentiation of Th1 and Th17 was decreased on flow cytometry analysis. Hematoxylin and eosin staining revealed reduced gastric mucosa inflammation, and enzyme-linked immunosorbent assay results demonstrated decreased levels of serum inflammatory cytokines. Furthermore, the terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL) results showed that DAPT treatment improved the apoptosis of gastric mucosal cells. Collectively, the findings indicate that Notch signaling is implicated in exacerbating H. pylori-induced inflammation by promoting macrophage activation and Th1/Th17 responses, highlighting its potential as a therapeutic target for alleviating the progression of H. pylori-related diseases.
Collapse
Affiliation(s)
- Yunxuan He
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Danli Xiao
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Hongfei Zhu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chuxi Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qiaoyuan Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jinling Xie
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Lvying Wei
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yueqi Dai
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.
| | - Yan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Li Z, Yu Y, Sun Q, Li Z, Huo X, Sha J, Qu D, Sun Y. Based on the TLR4/NLRP3 Pathway and Its Impact on the Formation of NETs to Explore the Mechanism of Ginsenoside Rg 1 on Acute Gouty Arthritis. Int J Mol Sci 2025; 26:4233. [PMID: 40362468 PMCID: PMC12071870 DOI: 10.3390/ijms26094233] [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: 03/26/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
This study investigated whether ginsenoside Rg1 (G-Rg1) alleviated acute gouty arthritis (AGA) in rats by modulating the TLR4/NLRP3 pathway and neutrophil extracellular trap (NET) formation. Rats were orally administered G-Rg1 or colchicine (Col) for 7 days, and monosodium urate (MSU) was injected into the ankle joints on day 5 to induce AGA. Joint swelling, histopathology (HE staining), and serum markers (MPO, NE, MPO-DNA, IL-6, IL-1β; ELISA) were assessed at the baseline and 6-36 h post-modeling. Western blot and immunofluorescence analyzed the NET-related and TLR4/NLRP3 pathway proteins in synovial tissue. G-Rg1 significantly reduced ankle swelling and synovial inflammation compared with the AGA group, lowered the serum IL-6, IL-1β, MPO, NE, and MPO-DNA levels, and suppressed NET-associated protein expression. Mechanistically, G-Rg1 downregulated TLR4/NLRP3 pathway activation in synovial tissue. These findings suggest that G-Rg1 mitigates AGA by inhibiting TLR4/NLRP3 signaling, thereby reducing inflammatory cytokine release and NET formation.
Collapse
Affiliation(s)
- Zhiman Li
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China or (Z.L.); (Y.Y.); (X.H.); (J.S.); (D.Q.)
| | - Yang Yu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China or (Z.L.); (Y.Y.); (X.H.); (J.S.); (D.Q.)
| | - Qiang Sun
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China;
| | - Zhilong Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 401300, China;
| | - Xiaohui Huo
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China or (Z.L.); (Y.Y.); (X.H.); (J.S.); (D.Q.)
| | - Jiyue Sha
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China or (Z.L.); (Y.Y.); (X.H.); (J.S.); (D.Q.)
| | - Di Qu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China or (Z.L.); (Y.Y.); (X.H.); (J.S.); (D.Q.)
| | - Yinshi Sun
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China;
| |
Collapse
|
19
|
Lv M, Zheng Y, Yuan M, Zhang E, Zheng M, Liu G, Zheng M, Gu W, Xu H. Cassiae semen extract ameliorates hyperlipidemia in rats by modulating lipid metabolism and FcγR-mediated immune regulation. Front Pharmacol 2025; 16:1546119. [PMID: 40357000 PMCID: PMC12066667 DOI: 10.3389/fphar.2025.1546119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Cassiae Semen Extract (CSE) shows promise in treating hyperlipidemia, although its underlying mechanisms are not yet fully understood. This study aimed to investigate the effects of CSE on hyperlipidemia in rats and explore the potential mechanisms involved. Methods Hyperlipidemic rats were induced by a high-fat diet (HFD) and treated with CSE. Serum, liver, and fecal samples were analyzed through biochemical assays, histopathological examination, 16S rRNA sequencing, KEGG pathway analysis, and Western blot. Results CSE treatment effectively alleviated biochemical imbalances and tissue damage induced by the HFD. 16S rRNA sequencing revealed that CSE improved gut microbiota dysbiosis and increased microbiota abundance. Pathological analysis showed that CSE reduced hepatic lipid accumulation, mitigating liver damage. KEGG pathway analysis suggested that the beneficial effects of CSE on hyperlipidemia may involve Fc gamma receptor (FcγR)-mediated phagocytosis, with immune activation influencing lipid homeostasis and liver inflammation. Western blot analysis further indicated that CSE may regulate lipid metabolism via Sterol Regulatory Element-Binding Protein-1c (SREBP-1c) and Peroxisome Proliferator-Activated Receptor Alpha (PPARα), while reducing hepatic inflammation through the MAPK signaling pathway. Discussion CSE may ameliorate hyperlipidemia in rats by modulating gut microbiota disorders, lipid metabolism, and FcγR-mediated immune regulation, providing a potential therapeutic approach for diseases associated with metabolic dysfunction and inflammation. However, further in-depth studies are required to fully elucidate these mechanisms.
Collapse
Affiliation(s)
- Mingyue Lv
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Yannan Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Man Yuan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Errui Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Zheng
- The Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangyao Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiliang Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongxi Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Gao J, Li W, Lin J, Han Y, Ji G, Liu Z. Galnt3, an enzyme engaged in protein glycosylation modification, is essential for the maintaining of intestinal health in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110373. [PMID: 40306380 DOI: 10.1016/j.fsi.2025.110373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/26/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Intestinal inflammation significantly impairs intestinal function and is closely associated with various health complications. Understanding its molecular mechanisms is crucial for developing effective therapeutic strategies. Galnt3, a member of the polypeptide N-acetylgalactosaminyltransferase family, participates in multiple biological processes, yet its specific role in intestinal inflammation remains poorly understood. In this study, we observed a significant downregulation of zebrafish galnt3 in response to GCRV virus or poly(I:C) infection. Galnt3 knockout (galnt3-/-) zebrafish exhibited reduced survival rates, particularly following GCRV virus inoculation, accompanied by severe ascites and abdominal hemorrhage. Histopathological examination of intestinal tissues revealed thinning of intestinal walls, shortened villi, and increased acidic mucus secretion, all indicative of aggravated intestinal inflammation. Furthermore, galnt3 deficiency was found to trigger the upregulation of numerous pro-inflammatory cytokine genes. Through cell scratch assays and p38 MAPK phosphorylation analysis, we demonstrated that Galnt3 inhibits p38 MAPK phosphorylation and macrophage migration, thereby reducing the production of pro-inflammatory factors. Our findings highlight the pivotal role of Galnt3 in maintaining intestinal homeostasis and regulating inflammatory responses, providing valuable insights into the molecular mechanisms underlying intestinal inflammation and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Jing Gao
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Wenjin Li
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Jingyuan Lin
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Yilin Han
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Guangdong Ji
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Zhenhui Liu
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
21
|
Park TJ, Choi BM, Hong H, Park JS, Kim SY. Anti-Inflammatory Effects of Ajuga decumbens Extract under Blue Light Stimulation. J Microbiol Biotechnol 2025; 35:e2502002. [PMID: 40295199 PMCID: PMC12089943 DOI: 10.4014/jmb.2502.02002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025]
Abstract
We investigated the potential of Ajuga decumbens Thunb. as an anti-inflammatory agent by utilizing plant resources to develop materials through the application of tissue culture and light-emitting diode (LED) cultivation technologies. To compare the changes between A. decumbens callus extract (ADCB) cultivated under blue monochromatic LED light and ADC (the negative control) cultivated under dark conditions, their morphological characteristics were tested and LC/MS analyses were conducted. ADCB exhibited a greenish hue compared with ADC and contained increased levels of specific compounds. The anti-inflammatory activities of the two samples were evaluated using LPS-stimulated macrophages. None of the samples exhibited cytotoxicity at any tested concentration. However, ADCB demonstrated a greater ability to reduce nitric oxide and key pro-inflammatory cytokines including interleukin-1β, interleukin-6, and tumor necrosis factor-α compared to the control. Furthermore, ADCB effectively suppressed the expression of inducible nitric oxide synthase and cyclooxygenase-2. It inhibited the phosphorylation of mitogen-activated protein kinase family proteins, including extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38, in a concentration-dependent manner. Tissue culture and LED cultivation technologies are significant methods for addressing plant supply challenges and enhancing the content of bioactive compounds, thereby increasing the applicability of plant materials. Moreover, ADCB produced using these technologies exhibited anti-inflammatory activity without causing irritation to human skin at active concentrations, suggesting its potential as a novel anti-inflammatory material.
Collapse
Affiliation(s)
- Tae-Jin Park
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| | - Byeong Min Choi
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| | - Hyehyun Hong
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| | - Jin-Soo Park
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Seung-Young Kim
- Department of Pharmaceutical Engineering & Biotechnology, Sunmoon University, Asan 31460, Republic of Korea
| |
Collapse
|
22
|
Yoo NH, Baek YS, Kim HK, Lee CO, Kim MJ. Antioxidant and Anti-Inflammatory Activities of Astilboides tabularis (Hemsl.) Engl. Root Extract. Molecules 2025; 30:1892. [PMID: 40363699 PMCID: PMC12073835 DOI: 10.3390/molecules30091892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Here, we examined the antioxidant and anti-inflammatory activities of the ethyl acetate (EtOAc) fraction of Astilboides tabularis (Hemsl.) Engl. root extracts, initially prepared from a 70% ethanol extraction. This EtOAc fraction exhibited significant scavenging activity against DPPH radicals (IC50: 11.38 ± 0.48 µg/mL) and ABTS radicals (IC50: 7.46 ± 0.58 µg/mL), and had a high total phenolic content (i.e., 407.02 ± 13.56 mg GAE/g). In addition, the EtOAc fraction demonstrated concentration-dependent protective effects in a RAW264.7 macrophage cell model subjected to oxidative stress. In lipopolysaccharide (LPS)-stimulated RAW264.7 cells, nitric oxide (NO) production and the expression of inflammatory mediators (iNOS, COX-2, TNF-α, IL-1β, IFN-β) were inhibited in a concentration-dependent manner. Western blot and real-time PCR (RT-PCR) analyses revealed that the EtOAc fraction also suppressed inflammatory mediator expression via inhibiting the activation of the NF-κB and MAPK signaling pathways. Finally, LC-QTOF-MS and LC-MS/MS analyses identified gallic acid and bergenin as compounds contributing to observed antioxidant and anti-inflammatory effects. In conclusion, the EtOAc fraction of A. tabularis root extracts exhibited strong anti-oxidant and anti-inflammatory properties, suggesting potential usage for treating various inflammatory diseases.
Collapse
Affiliation(s)
- Nam Ho Yoo
- Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.H.Y.); (Y.S.B.)
| | - Young Sun Baek
- Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.H.Y.); (Y.S.B.)
| | - Hee Kyu Kim
- Gangwondo Forest Science Institute, Chuncheon 24207, Republic of Korea; (H.K.K.); (C.O.L.)
| | - Chan Ok Lee
- Gangwondo Forest Science Institute, Chuncheon 24207, Republic of Korea; (H.K.K.); (C.O.L.)
| | - Myong Jo Kim
- Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (N.H.Y.); (Y.S.B.)
| |
Collapse
|
23
|
Albini A, Di Paola L, Mei G, Baci D, Fusco N, Corso G, Noonan D. Inflammation and cancer cell survival: TRAF2 as a key player. Cell Death Dis 2025; 16:292. [PMID: 40229245 PMCID: PMC11997178 DOI: 10.1038/s41419-025-07609-w] [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: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
TNF receptor-associated factor 2 (TRAF2) plays a crucial role in both physiological and pathological processes. It takes part in the regulation of cell survival and death, tissue regeneration, development, endoplasmic reticulum stress response, autophagy, homeostasis of the epithelial barrier and regulation of adaptive and innate immunity. Initially identified for its interaction with TNF receptor 2 (TNFR2), TRAF2 contains a TRAF domain that enables homo- and hetero-oligomerization, allowing it to interact with multiple receptors and signaling molecules. While best known for mediating TNFR1 and TNFR2 signaling, TRAF2 also modulates other receptor pathways, including MAPK, NF-κB, and Wnt/β-catenin cascades. By regulating NF-κB-inducing kinase (NIK), TRAF2 is a key activator of the alternative NF-κB pathway, linking it to inflammatory diseases, immune dysfunction, and tumorigenesis. In the innate immune system, TRAF2 influences macrophage differentiation, activation, and survival and stimulates natural killer cell cytotoxicity. In the adaptive immune system, it represses effector B- and T-cell activity while sustaining regulatory T-cell function, thus promoting immune suppression. The lack of fine-tuning of TRAF2 activity leads to excessive NF-kB activation, driving chronic inflammation and autoimmunity. Although TRAF2 can act as a tumor suppressor, it is predominantly described as a tumor promoter, as its expression has been correlated with increased metastatic potential and poorer prognosis in several types of cancer. Targeting TRAF2 or TRAF2-dependent signaling pathways might represent a promising anti-cancer therapeutic strategy.
Collapse
Grants
- The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- PRIN 2022, grant 2022PJKF88 The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- PRIN 2022 The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- "Umberto Veronesi" Foundation project: "Massive CDH1 genetic screening in the so-called hereditary breast-gastric cancer syndrome". The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
Collapse
Affiliation(s)
- Adriana Albini
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
| | - Luisa Di Paola
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Faculty Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico, Rome, Italy
| | - Giampiero Mei
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Denisa Baci
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan, Italy
| | - Nicola Fusco
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giovanni Corso
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
- Division of Breast Surgery, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
| | - Douglas Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
24
|
Zhang M, Feng C, Zhang B, Yin Y, Chen J, Liu H, Farag MA, Mamadalieva NZ, Li N, Sun J, Sun S, Liu C. In vitro and in vivo immune-enhancing effects of punicic acid and the action mechanisms as revealed via microbiome and lipid profiling. Food Funct 2025; 16:3120-3133. [PMID: 40159912 DOI: 10.1039/d4fo05023a] [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: 04/02/2025]
Abstract
Punicic acid (PA) is a chief component of pomegranate seed oil with several health benefits. In this study, the in vitro immunomodulatory activity of PA was assessed using RAW264.7 cells, revealing that PA activated the macrophages, facilitated the concentration of immune-related cytokines and enzymes, and regulated the immune-related NF-κB and MAPK signaling pathways. Further, the in vivo immune-enhancing effect of PA was evaluated with the cyclophosphamide (CTX)-induced immune-compromised mouse model with 16S rDNA amplicon sequencing and relative quantification of lipidome. Results indicated that high doses of PA (200 mg kg-1) remarkably restored CTX-induced immune injury by enhancing the innate and adaptive immunity to stimulate the secretion of immune-related factors. In addition, PA improved gut microbiota dysbiosis and ameliorated lipid metabolism disorders. Our research provides a theoretical basis for the exploitation of PA as a functional component with immune-enhancing effects and adds to the potential health uses of pomegranate seed oil.
Collapse
Affiliation(s)
- Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Caiyun Feng
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China
| | - Bo Zhang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China
| | - Yanlei Yin
- Shandong Institute of Pomology, Tai'an, 271000, China
| | - Jinlong Chen
- Work Station of Forest Fruit Industry in Kashi, Kashi, 844000, PR China
| | - Haoran Liu
- JiMei One Health Industry (Shandong) Co., Ltd, Zaozhuang, 277300, PR China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nilufar Z Mamadalieva
- Institute of the Chemistry of Plant Substances of the Academy Sciences of Uzbekistan, Tashkent, 100170, Uzbekistan
| | - Ningyang Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, PR China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Shutao Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- Shandong Aojing Biotechnology Co., Ltd, Jining, 273500, PR China
| |
Collapse
|
25
|
Manivasagam S, Han J, Teghanemt A, Keen H, Sownthirarajan B, Cheng B, Singh A, Lewis A, Vogel OA, Loganathan G, Huang L, Panis M, Meyerholz DK, tenOever B, Perez JT, Manicassamy S, Issuree PD, Manicassamy B. Transcriptional repressor Capicua is a gatekeeper of cell-intrinsic interferon responses. Cell Host Microbe 2025; 33:512-528.e7. [PMID: 40132591 PMCID: PMC11985295 DOI: 10.1016/j.chom.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/27/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025]
Abstract
Early detection of viral infection and rapid activation of host antiviral defenses through transcriptional upregulation of interferons (IFNs) and IFN-stimulated genes (ISGs) are critical for controlling infection. However, aberrant production of IFN in the absence of viral infection leads to auto-inflammation and can be detrimental to the host. Here, we show that the DNA-binding transcriptional repressor complex composed of Capicua (CIC) and Ataxin-1 like (ATXN1L) binds to an 8-nucleotide motif near IFN and ISG promoters and prevents erroneous expression of inflammatory genes under homeostasis in humans and mice. By contrast, during respiratory viral infection, activation of the mitogen-activated protein kinase (MAPK) pathway results in rapid degradation of the CIC-ATXN1L complex, thereby relieving repression and allowing for robust induction of IFN and ISGs. Together, our studies define a new paradigm for host regulation of IFN and ISGs through the evolutionarily conserved CIC-ATXN1L transcriptional repressor complex during homeostasis and viral infection.
Collapse
Affiliation(s)
| | - Julianna Han
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Athmane Teghanemt
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Henry Keen
- Bioinformatics Division of the Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA
| | | | - Boyang Cheng
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Abhiraj Singh
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Abigail Lewis
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Olivia A Vogel
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Gayathri Loganathan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lei Huang
- Center for Research Informatics, The University of Chicago, Chicago, IL 60637, USA
| | - Maryline Panis
- Department of Microbiology, New York University, New York, NY, USA
| | | | | | - Jasmine T Perez
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | | | - Priya D Issuree
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
26
|
Liu C, Liu X, Duan J. Artemisinin and Its Derivatives: Promising Therapeutic Agents for Age-Related Macular Degeneration. Pharmaceuticals (Basel) 2025; 18:535. [PMID: 40283970 PMCID: PMC12030120 DOI: 10.3390/ph18040535] [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: 03/09/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment and blindness in older adults. Its pathogenesis involves multiple factors, including aging, environmental influences, genetic predisposition, oxidative stress, metabolic dysfunction, and immune dysregulation. Currently, AMD treatment focuses primarily on wet AMD, managed through repeated intravitreal injections of anti-vascular endothelial growth factor (VEGF) therapies. While anti-VEGF agents represent a major breakthrough in wet AMD care, repeated injections may lead to incomplete responses or resistance in some patients, and carry a risk of progressive fibrosis. Artemisinin (ART) and its derivatives, originally developed as antimalarial drugs, exhibit a broad spectrum of pleiotropic activities beyond their established use, including anti-inflammatory, anti-angiogenic, antioxidant, anti-fibrotic, mitochondrial regulatory, lipid metabolic, and immunosuppressive effects. These properties position ART as a promising therapeutic candidate for AMD. A growing interest in ART-based therapies for AMD has emerged in recent years, with numerous studies demonstrating their potential benefits. However, no comprehensive review has systematically summarized the specific roles of ART and its derivatives in AMD pathogenesis and treatment. This paper aims to fill the knowledge gap by synthesizing the therapeutic efficacy and molecular mechanisms of ART and its derivatives in AMD, thereby providing a foundation for future investigations.
Collapse
Affiliation(s)
- Chun Liu
- Eye School, Chengdu University of TCM, Chengdu 610075, China
| | - Xiaoqin Liu
- Clinical Medical School, Chengdu University of TCM, Chengdu 610075, China
| | - Junguo Duan
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu 610075, China
| |
Collapse
|
27
|
Li K, Ji X, Tian S, Li J, Tian Y, Ma X, Li H, Zhang H, Chen CT, Gu W. Oxidative stress in asthma pathogenesis: mechanistic insights and implications for airway smooth muscle dysfunction. Cell Tissue Res 2025; 400:17-34. [PMID: 39918765 DOI: 10.1007/s00441-025-03953-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/28/2025] [Indexed: 04/04/2025]
Abstract
Airway smooth muscle (ASM) dysfunction is a key factor in the narrowing of airways in asthma patients, characterized by excessive secretion of inflammatory factors, increased mass, and amplified contractile responses. These pathological features are instrumental in the propagation of airway inflammation, structural remodeling, and the escalation of airway hyperresponsiveness (AHR), which are also principal factors underlying the limitations of current therapeutic strategies. In asthmatic ASM, an imbalance between oxidant production and antioxidant defenses culminates in oxidative stress, which is involved in the excessive secretion of inflammatory factors, increased mass, and amplified contractile responses of ASM, and is a critical etiological factor implicated in the dysregulation of ASM function. The molecular pathways through which oxidative stress exerts its effects on ASM in asthma are multifaceted, with the Nrf2/HO-1, MAPK, and PI3K/Akt pathways being particularly noteworthy. These characteristic pathways play a potential role by connecting with different upstream and downstream signaling molecules and are involved in the amplification of ASM inflammatory responses, increased mass, and AHR. This review provides a comprehensive synthesis of the phenotypic expression of ASM dysfunction in asthma, the interplay between oxidants and antioxidants, and the evidence base and molecular underpinnings linking oxidative stress to ASM dysfunction. Given the profound implications of ASM dysfunction on the airflow limitation in asthma and the seminal role of oxidative stress in this process, a deeper exploration of these mechanisms is essential for unraveling the pathogenesis of asthma and may offer novel perspectives for its prophylaxis and management.
Collapse
Affiliation(s)
- Kangxia Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Xiang Ji
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Shan Tian
- College of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Jian Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Yizhu Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Xiaoqing Ma
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Huanping Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Hong Zhang
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, People's Republic of China
| | - Cai-Tao Chen
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, People's Republic of China.
| | - Wei Gu
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China.
| |
Collapse
|
28
|
He X, He Y, Deng X, Lu N, Li A, Gao S, He S, Wang Y, Fu N, Wang Z, Nie Y, Xu L. Rv2741 Promotes Mycobacterium Survival by Modulating Macrophage Function via the IL-1α-MAPK Axis. ACS Infect Dis 2025; 11:676-688. [PMID: 40009799 DOI: 10.1021/acsinfecdis.4c00790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
One of the primary healthcare problems in the world today is tuberculosis (TB), a chronic infectious illness brought on by Mycobacterium tuberculosis (M. tuberculosis). A distinct family of PE_PGRS proteins, encoded by the M. tuberculosis genome, has attracted more attention because of their involvement in immune evasion and bacterial pathogenicity. Nevertheless, the specific functions and mechanisms of action for the majority of PE_PGRS proteins remain largely unexplored. This study focuses on the Rv2741 (PE_PGRS47) gene, which is exclusively present in pathogenic mycobacteria. To examine the function of Rv2741 in host-pathogen interactions, we created recombinant strains of Mycobacterium smegmatis (M. smegmatis) that expressed the M. tuberculosis Rv2741 gene. IL-1α was found to be a key mediator of host response modulation by Rv2741. Rv2741 downregulates the secretion of IL-1α and inhibits the MAPK signaling pathway, particularly the p38 and ERK1/2 pathways, thereby cooperatively inhibiting macrophage autophagy and apoptosis. Meanwhile, the decrease in IL-1α secretion directly leads to changes in the cytokine secretion pattern and a reduction in nitric oxide (NO) production. This multifaceted regulatory mechanism ultimately favors the survival of M. smegmatis in macrophages. This research significantly expands our understanding of Rv2741 function, revealing its crucial role as a multifunctional virulence factor in the immune evasion of M. tuberculosis.
Collapse
Affiliation(s)
- Xintong He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yonglin He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xichuan Deng
- Pathogen Biology and Immunology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Nan Lu
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Anlong Li
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Sijia Gao
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shiyan He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuran Wang
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Nanzhe Fu
- International Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Zijie Wang
- International Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Yuxin Nie
- The Second Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Lei Xu
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
29
|
Han HJ, Hyun CG. Anti-Inflammatory Effects and Human Skin Safety of the Eastern Traditional Herb Mosla japonica. Life (Basel) 2025; 15:418. [PMID: 40141763 PMCID: PMC11943674 DOI: 10.3390/life15030418] [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: 01/21/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Traditional knowledge has long provided natural solutions for disease prevention and treatment, complementing modern medicine. Mosla japonica (Korean mint) has been traditionally valued for its pesticidal, dehumidifying, anti-swelling, and detoxifying properties. This study explores its anti-inflammatory potential using M. japonica extract (MJE) in LPS-stimulated RAW 264.7 macrophages and evaluates its safety for human skin applications. MJE significantly reduced inflammatory mediators such as nitric oxide (NO), prostaglandin E2 (PGE2), and key cytokines (IL-1β, IL-6, TNF-α) in a dose-dependent manner. It also suppressed the expression of iNOS and COX-2, enzymes crucial for inflammation. Mechanistically, MJE inhibited NF-κB activation by stabilizing IκBα, thereby reducing inflammation-related gene expression. Additionally, it downregulated ERK, JNK, and p38 in the MAPK signaling pathway, further contributing to its anti-inflammatory effects. A primary skin irritation test confirmed MJE's safety, showing no significant skin reactions at 100 μg/mL. These findings highlight MJE's strong anti-inflammatory properties and potential for dermatological applications. This study underscores the pharmacological value of M. japonica and its integration into modern scientific research, aligning with global biodiversity frameworks such as the Nagoya Protocol. Future research may further expand its applications in medicine and skincare.
Collapse
Affiliation(s)
| | - Chang-Gu Hyun
- Jeju Inside Agency and Cosmetic Science Center, Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Republic of Korea;
| |
Collapse
|
30
|
Huang K, Zhang Q, Wan H, Ban X, Chen X, Wan X, Lu R, He Y, Xiong K. TAK1 at the crossroads of multiple regulated cell death pathways: from molecular mechanisms to human diseases. FEBS J 2025. [DOI: 10.1111/febs.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
Regulated cell death (RCD), the form of cell death that can be genetically controlled by multiple signaling pathways, plays an important role in organogenesis, tissue remodeling, and maintenance of organism homeostasis and is closely associated with various human diseases. Transforming growth factor‐beta‐activated kinase 1 (TAK1) is a member of the serine/threonine protein kinase family, which can respond to different internal and external stimuli and participate in inflammatory and immune responses. Emerging evidence suggests that TAK1 is an important regulator at the crossroad of multiple RCD pathways, including apoptosis, necroptosis, pyroptosis, and PANoptosis. The regulation of TAK1 affects disease progression through multiple signaling pathways, and therapeutic strategies targeting TAK1 have been proposed for inflammatory diseases, central nervous system diseases, and cancers. In this review, we provide an overview of the downstream signaling pathways regulated by TAK1 and its binding proteins. Their critical regulatory roles in different forms of cell death are also summarized. In addition, we discuss the potential of targeting TAK1 in the treatment of human diseases, with a specific focus on neurological disorders and cancer.
Collapse
Affiliation(s)
- Kun Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Xiangya School of Medicine Central South University Changsha China
| | - Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Department of Ophthalmology Stanford University School of Medicine Palo Alto CA USA
- Key Laboratory of Emergency and Trauma of Ministry of Education, College of Emergency and Trauma Hainan Medical University Haikou China
| | - Hao Wan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xiao‐Xia Ban
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xin‐Yu Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xin‐Xing Wan
- Department of Endocrinology Third Xiangya Hospital, Central South University Changsha China
| | - Rui Lu
- Department of Molecular and Cellular Physiology Stanford University Stanford CA USA
| | - Ye He
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Changsha Aier Eye Hospital China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Key Laboratory of Emergency and Trauma of Ministry of Education, College of Emergency and Trauma Hainan Medical University Haikou China
- Hunan Key Laboratory of Ophthalmology Changsha China
| |
Collapse
|
31
|
Ma L, Xie L, Wu Q, Jin L, Li J, Tang L, Zhang L, Chen L, Qiu Z. Targeting the S100A9/P38 MAPK/HSPB1 axis as a novel approach for aortic dissection therapy. Int Immunopharmacol 2025; 149:114225. [PMID: 39904041 DOI: 10.1016/j.intimp.2025.114225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/02/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Aortic dissection (AD) is caused by inflammatory responses and extracellular matrix (ECM) degradation processes, in which S100A9, a proinflammatory protein, may play a role. This study explored the role S100A9/P38 MAPK/HSPB1 signaling axis in AD pathogenesis and the therapeutic potential of targeting this pathway. METHODS S100A9 expression in the aortic tissues of patients with AD/healthy controls were analyzed using bioinformatics, ELISA, qPCR, western blotting, and immunohistochemistry. In an AD mouse model induced by β-aminopropionitrile and angiotensin II (Ang-II), S100A9 expression was inhibited using specific inhibitors to assess its relationship with AD, and proteomics were performed to explore the pathways related to S100A9 expression. Human aortic vascular smooth muscle cells (HVSMC) were treated with Ang-II, S100A9 knockdown, P38 MAPK inhibitors, and HSPB1 knockdown, and experimental methods were used to assess changes in inflammatory cytokines, ECM remodeling, cell proliferation, and apoptosis. Rescue experiments validated the role of the S100A9/P38 MAPK/HSPB1 axis. RESULTS S100A9 was significantly upregulated in patients with AD, while levels of inflammatory cytokines and matrix metalloproteinases (MMPs) were elevated. S100a9 inhibition reduced the incidence of AD, improved survival, and stabilized the aortic structure in mice, with reduced collagen deposition and SMC apoptosis in vitro. S100A9 knockdown reduces Ang-II-induced HVSMC proliferation, apoptosis resistance, and ECM degradation. Mechanistic studies revealed that the S100A9/P38 MAPK/HSPB1 axis regulates inflammatory cytokine and MMPs release. CONCLUSION S100A9 regulates inflammation and ECM degradation through the P38 MAPK/HSPB1 axis, influencing HVSMC proliferation and apoptosis and promoting AD development. This pathway may be a promising therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Likang Ma
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Linfeng Xie
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Qingsong Wu
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Lei Jin
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Jiakang Li
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences Fujian Medical University Fuzhou China
| | - Lele Tang
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Li Zhang
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences Fujian Medical University Fuzhou China.
| | - Liangwan Chen
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China.
| | - Zhihuang Qiu
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China.
| |
Collapse
|
32
|
Abou Mansour M, El Rassi C, Sleem B, Borghol R, Arabi M. Thromboembolic Events in the Era of COVID-19: A Detailed Narrative Review. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:3804576. [PMID: 40226433 PMCID: PMC11986918 DOI: 10.1155/cjid/3804576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is not only characterized by respiratory symptoms but is also associated with a wide range of systemic complications, including significant hematologic abnormalities. This is a comprehensive review of the current literature, using PubMed and Google Scholar, on the pathophysiology and incidence of thromboembolic events in COVID-19 patients and thromboprophylaxis. COVID-19 infection induces a prothrombotic state in patients through the dysregulation of the renin-angiotensin-aldosterone system (RAAS), endothelial dysfunction, elevated von Willebrand factor (vWF), and a dysregulated immune response involving the complement system and neutrophil extracellular traps (NETs). As a result, thromboembolic complications have emerged in COVID-19 cases, occurring more frequently in severe cases and hospitalized patients. These thrombotic events affect both venous and arterial circulation, with increased incidences of deep venous thrombosis (DVT), pulmonary embolism (PE), systemic arterial thrombosis, and myocardial infarction (MI). While DVT and PE are more common, the literature highlights the potential lethal consequences of arterial thromboembolism (ATE). This review also briefly examines the ongoing discussions regarding the use of anticoagulants for the prevention of thrombotic events in COVID-19 patients. While theoretically promising, current studies have yielded varied outcomes: Some suggest potential benefits, whereas others report an increased risk of bleeding events among hospitalized patients. Therefore, further large-scale studies are needed to assess the efficacy and safety of anticoagulants for thromboprophylaxis in COVID-19 patients.
Collapse
Affiliation(s)
- Maria Abou Mansour
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Christophe El Rassi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Bshara Sleem
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raphah Borghol
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
33
|
Yin L, Wang H, Xu H, Lu H, Lv J, Lu C. Asperuloside suppresses the progression of depression through O-GlcNAcylation of IκBα and regulating NFκB signaling. J Pharmacol Sci 2025; 157:179-188. [PMID: 39929592 DOI: 10.1016/j.jphs.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 05/08/2025] Open
Abstract
Depression is a pervasive mental disorder that poses a significant threat to human health globally. Asperuloside (ASP), an iridoid glycoside extracted from Herba Paederiae, exhibits a range of pharmacological activities, including anti-tumor and anti-inflammatory effects. This study aims to explore the function and molecular mechanisms of ASP in alleviating depression. Chronic unpredictable mild stress (CUMS) was employed to establish a rat model of depression. Behavioral tests were conducted to evaluate the antidepressant effects of ASP. Apoptosis in hippocampal tissues was assessed using TUNEL assay. Primary hippocampal neuron apoptosis was assessed using Annexin V/PI staining and flow cytometry, while cell death was detected via PI staining. The expression levels of target mRNAs and proteins were analyzed by quantitative PCR (qPCR) and western blotting, respectively. Additionally, the levels of O-GlcNAcylation and ubiquitination were determined by western blot analysis following immunoprecipitation. Molecular docking was performed to elucidate the interaction mode between ASP and its target protein, O-linked β-N-acetylglucosamine transferase (OGT). Our findings revealed that ASP treatment significantly ameliorated depression-like behaviors and cognitive dysfunction, as well as inhibited hippocampus apoptosis in CUMS-induced rats, Moreover, ASP inhibited LPS-induced neuronal cell apoptosis and suppressed the activation of the NF-κB signaling pathway. Mechanistically, we demonstrated that ASP promoted O-GlcNAcylation of IκBα, and suppressed its ubiquitination and phosphorylation, thereby stabilizing IκBα protein. In conclusion, ASP exerts antidepressant effects by enhancing IκBα O-GlcNAcylation, thus inhibiting its ubiquitination and phosphorylation. These findings provide a novel therapeutic target for the treatment of depression.
Collapse
Affiliation(s)
- Li Yin
- Zunyi Medical and Pharmaceutical College, No. 8, North Section of Ping'an Avenue, Honghuagang District, Zunyi City, Guizhou, China
| | - Huakun Wang
- Zunyi Medical and Pharmaceutical College, No. 8, North Section of Ping'an Avenue, Honghuagang District, Zunyi City, Guizhou, China
| | - Hong Xu
- Department of Scientific Research and Technology, Xinjiang Uyghur Autonomous Region Institute for Drug Control, No.518, Xiba Jiahu Road, Xinshi District, Urumqi, 830054, Xinjiang, China
| | - Haixiao Lu
- Department of Biopharmaceutics, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China; Bioengineering & Technology Center for Native Medicinal Resources Development, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China
| | - Jiayu Lv
- Department of Biopharmaceutics, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China
| | - Chengshu Lu
- Department of Biopharmaceutics, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China.
| |
Collapse
|
34
|
Veinstein M, Stroobant V, Wavreil F, Michiels T, Sorgeloos F. The "DDVF" motif used by viral and bacterial proteins to hijack RSK kinases mimics a short linear motif (SLiM) found in proteins related to the RAS-ERK MAP kinase pathway. PLoS Pathog 2025; 21:e1013016. [PMID: 40153681 PMCID: PMC11984722 DOI: 10.1371/journal.ppat.1013016] [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: 08/08/2024] [Revised: 04/10/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025] Open
Abstract
Proteins of pathogens such as cardioviruses, Kaposi sarcoma-associated herpes virus, varicella zoster virus and bacteria of the genus Yersinia were previously shown to use a common "DDVF" (D/E-D/E-V-F) short linear motif (SLiM) to hijack cellular kinases of the RSK (p90 ribosomal S6 kinases) family. Notably, the leader (L) protein of Theiler's murine encephalomyelitis virus (TMEV), a cardiovirus, and protein YopM of Yersinia species were shown to act as adapters to retarget RSKs toward unconventional substrates, nucleoporins and pyrin, respectively. Remarkable conservation of the SLiM docking site targeted by pathogens' proteins in RSK sequences suggested a physiological role for this site. Using SLiM prediction tools and AlphaFold docking, we screened the human proteome for proteins that would interact with RSKs through a DDVF-like SLiM. Co-immunoprecipitation experiments show that two candidates previously known as RSK partners, FGFR1 and SPRED2, as well as two candidates identified as novel RSK partners, GAB3 and CNKSR2 do interact with RSKs through a similar interface as the one used by pathogens, as was recently documented for SPRED2. FGFR1 employs a DSVF motif to bind RSKs and phosphorylation of the serine in this motif slightly increased RSK binding. FGFR1, SPRED2, GAB3 and CNKSR2 act upstream of RSK in the RAS-ERK MAP kinase pathway. Analysis of ERK activation in cells expressing a mutated form of RSK lacking the DDVF-docking site suggests that RSK might interact with the DDVF-like SLiM of several partners to provide a negative feed-back to the ERK MAPK pathway. Moreover, after TMEV infection, ERK phosphorylation was altered by the L protein in a DDVF-dependent manner. Taken together, our data suggest that, in addition to retargeting RSKs toward unconventional substrates, pathogens' proteins carrying a DDVF-like motif can compete with endogenous DDVF-containing proteins for RSK binding, thereby altering the regulation of the RAS-ERK MAP kinase pathway.
Collapse
Affiliation(s)
- Martin Veinstein
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | - Fanny Wavreil
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Thomas Michiels
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Sorgeloos
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
35
|
Zhong C, Li W, Zhang X, Zhang D, Wen Z, Song W, Jiang Z, Gao Z, Guo H, Bi G, Liu Z, Li D, Dinesh-Kumar SP, Zhang Y. A cell wall-associated kinase phosphorylates NLR immune receptor to negatively regulate resistosome formation. NATURE PLANTS 2025; 11:561-579. [PMID: 40119183 DOI: 10.1038/s41477-025-01949-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/21/2025] [Indexed: 03/24/2025]
Abstract
Plants deploy intracellular nucleotide-binding leucine-rich repeats (NLRs) to detect pathogen effectors and initiate immune responses. Although the activation mechanism of some plant NLRs forming resistosomes has been elucidated, whether NLR resistosome assembly is regulated to fine-tune immunity remains enigmatic. Here we used an antiviral coiled coil-nucleotide-binding site-leucine rich repeat, Barley Stripe Resistance 1 (BSR1), as a model and demonstrate that BSR1 is phosphorylated. Using a proximity labelling approach, we identified a wall-associated kinase-like protein 20 (WAKL20) which negatively regulates BSR1-mediated immune responses by directly phosphorylating the Ser470 residue in the NB-ARC domain of BSR1. Mechanistically, Ser470 phosphorylation results in a steric clash of intramolecular domains of BSR1, thereby compromising BSR1 oligomerization. The phosphorylation site is conserved among multiple plant NLRs and our results show that WAKL20 participates in other NLR-mediated immune responses besides BSR1. Together, our data reveal phosphorylation as a mechanism for modulating plant resistosome assembly, and provide new insight into NLR-mediated plant immunity.
Collapse
Affiliation(s)
- Chenchen Zhong
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wenli Li
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xinyu Zhang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Dingliang Zhang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhiyan Wen
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wen Song
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhihao Jiang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zongyu Gao
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hailong Guo
- Ministry of Agriculture Key Laboratory of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Guozhi Bi
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhiyong Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, The Innovative Academy of Seed Design, Chinese Academy of Science, Beijing, China
| | - Dawei Li
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Savithramma P Dinesh-Kumar
- Department of Plant Biology and The Genome Center, College of Biological Sciences, University of California, Davis, Davis, CA, USA
| | - Yongliang Zhang
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
36
|
Finnerty RM, Carulli DJ, Hedge A, Wang Y, Boadu F, Winuthayanon S, Jack Cheng J, Winuthayanon W. Multi-omics analyses and machine learning prediction of oviductal responses in the presence of gametes and embryos. eLife 2025; 13:RP100705. [PMID: 40009070 PMCID: PMC11864756 DOI: 10.7554/elife.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
The oviduct is the site of fertilization and preimplantation embryo development in mammals. Evidence suggests that gametes alter oviductal gene expression. To delineate the adaptive interactions between the oviduct and gamete/embryo, we performed a multi-omics characterization of oviductal tissues utilizing bulk RNA-sequencing (RNA-seq), single-cell RNA-sequencing (scRNA-seq), and proteomics collected from distal and proximal at various stages after mating in mice. We observed robust region-specific transcriptional signatures. Specifically, the presence of sperm induces genes involved in pro-inflammatory responses in the proximal region at 0.5 days post-coitus (dpc). Genes involved in inflammatory responses were produced specifically by secretory epithelial cells in the oviduct. At 1.5 and 2.5 dpc, genes involved in pyruvate and glycolysis were enriched in the proximal region, potentially providing metabolic support for developing embryos. Abundant proteins in the oviductal fluid were differentially observed between naturally fertilized and superovulated samples. RNA-seq data were used to identify transcription factors predicted to influence protein abundance in the proteomic data via a novel machine learning model based on transformers of integrating transcriptomics and proteomics data. The transformers identified influential transcription factors and correlated predictive protein expressions in alignment with the in vivo-derived data. Lastly, we found some differences between inflammatory responses in sperm-exposed mouse oviducts compared to hydrosalpinx Fallopian tubes from patients. In conclusion, our multi-omics characterization and subsequent in vivo confirmation of proteins/RNAs indicate that the oviduct is adaptive and responsive to the presence of sperm and embryos in a spatiotemporal manner.
Collapse
Affiliation(s)
- Ryan M Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-ColumbiaColumbiaUnited States
| | - Daniel J Carulli
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| | - Akshata Hedge
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Yanli Wang
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Frimpong Boadu
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| | - Jianlin Jack Cheng
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Wipawee Winuthayanon
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-ColumbiaColumbiaUnited States
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| |
Collapse
|
37
|
Tie X, Chen Z, Yao S, Wu B, Yan B, Zhai H, Qiao X, Su X, Wang L. Immune Imbalance in Primary Membranous Nephropathy at Single-cell Resolution. FRONT BIOSCI-LANDMRK 2025; 30:36332. [PMID: 40018947 DOI: 10.31083/fbl36332] [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: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/25/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Primary membranous nephropathy (pMN) often progresses to end-stage renal disease (ESRD) in the absence of immunosuppressive therapy. The immunological mechanisms driving pMN progression remain insufficiently understood. METHODS We developed a single-cell transcriptomic profile of peripheral blood mononuclear cells (PBMCs) from 11 newly-diagnosed pMN patients and 5 healthy donors. Through correlation analysis, we identified potential biomarkers for disease stratification and poor prognosis. RESULTS Expression levels of several proinflammatory factors were significantly increased in patients compared to healthy donors, such as interleukins (IL1B, IL8, and IL15) and interferon G (IFNG). Multiple pattern recognition receptors involved in proinflammatory signaling were also upregulated in patients, including NOD-like receptors (NLRs) (NLRP1, NLRP3, and NLRC5), RNA helicases (DDX58, IFIH1, DHX9, and DHX36), cGAS (cyclic GMP-AMP synthase) and IFI16 (interferon gamma inducible protein 16). Additionally, human leukocyte antigen molecules HLA-DQA1 and HLA-DRB1 enriched in memory B cells were upregulated in patients. More importantly, we found that the genes for antiviral defense response were significantly elevated in high-risk patients relative to the low-risk group. More than twenty genes were negatively correlated with estimated glomerular filtration rate (eGFR), such as BST2 (bone marrow stromal cell antigen 2) and SLC35F1 (solute carrier family 35 member F1). Their predicted values were confirmed in a larger population with nephrotic syndrome or other chronic kidney diseases from a public database. Furthermore, we developed a series of scoring systems for distinguishing high-risk patients from low- and moderate-risk individuals. CONCLUSIONS Our study provides insight into the immunological mechanism of pMN and identifies numerous biomarkers and signaling pathways as potential therapeutic targets for managing the progression of high-risk pMN.
Collapse
Affiliation(s)
- Xuan Tie
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Zhiang Chen
- Zhejiang University School of Medicine, 310058 Hangzhou, Zhejiang, China
| | - Shulei Yao
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Binxin Wu
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Bingjuan Yan
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Huifang Zhai
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Xi Qiao
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Xiaole Su
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| | - Lihua Wang
- Department of Nephrology, Second Hospital of Shanxi Medical University, 030000 Taiyuan, Shanxi, China
- Shanxi Kidney Disease Institute, 030000 Taiyuan, Shanxi, China
- Institute of Nephrology, Shanxi Medical University, 030000 Taiyuan, Shanxi, China
| |
Collapse
|
38
|
Haacke N, Wang H, Yan S, Barovic M, Li X, Nagai K, Botezatu A, Hatzioannou A, Gercken B, Trimaglio G, Shah AU, Wang J, Ye L, Jaykar MT, Rauner M, Wielockx B, Chung KJ, Netea MG, Kalafati L, Hajishengallis G, Chavakis T. Innate immune training of osteoclastogenesis promotes inflammatory bone loss in mice. Dev Cell 2025:S1534-5807(25)00063-2. [PMID: 40020679 PMCID: PMC7617534 DOI: 10.1016/j.devcel.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
We previously demonstrated that long-term trained immunity (TRIM) involves adaptations that imprint innate immune memory in long-lived myelopoiesis precursors and their progeny, monocytes/macrophages and neutrophils, which thereby acquire enhanced responsiveness to future challenges. Here, we show that a distinct component of myeloid biology, osteoclastogenesis, can also undergo innate immune training. Indeed, β-glucan-induced TRIM was associated with an increased osteoclastogenesis bias in the bone marrow and an expansion of monocytes/osteoclast progenitors in the periphery, resulting in aggravated severity of experimental periodontitis and arthritis. In the setting of trained inflammatory osteoclastogenesis, we observed transcriptomic rewiring in synovial myeloid cells of arthritic mice, featuring prominent upregulation of the transcription factor melanogenesis-associated transcription factor (MITF). Adoptive transfer of splenic monocytes from β-glucan-trained mice to naive recipients exacerbated arthritis in the latter in a strictly MITF-dependent manner. Our findings establish trained osteoclastogenesis as a maladaptive component of TRIM and potentially provide therapeutic targets in inflammatory bone loss disorders.
Collapse
Affiliation(s)
- Nora Haacke
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shu Yan
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Marko Barovic
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kosuke Nagai
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Adelina Botezatu
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Aikaterini Hatzioannou
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Giulia Trimaglio
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Anisha U Shah
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mangesh T Jaykar
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 XZ Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| |
Collapse
|
39
|
Ha B, Kang JH, Kim DH, Lee MY. Lipopolysaccharide-Induced Inflammatory Response and Its Prominent Suppression by Paspalum thunbergii Extract. Int J Mol Sci 2025; 26:1611. [PMID: 40004077 PMCID: PMC11855676 DOI: 10.3390/ijms26041611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The extract of Paspalum thunbergii, a native perennial herb in Korea belonging to the rice family, was investigated for its anti-inflammatory activity and the underlying mechanisms driving its effects. Fifteen chemical components of the P. thunbergii extract, including rosmarinic acid and isoquercitrin, were identified using LC-MS. The extract showed antioxidative activity through DPPH and ABTS cation radical scavenging activity. The P. thunbergii extract significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in macrophage RAW 264.7 cells. The extract inhibited the expression of lipopolysaccharide-induced iNOS and COX-2, which are inflammation-related enzymes. To explore the underlying anti-inflammatory mechanism, the expression levels of signal proteins related to MAPK, NF-κB, JAK/STAT, and Wnt/β-catenin signaling were measured. As a result, the P. thunbergii extract inhibited the expression of p-p38, and p-JNK increased by LPS in RAW 264.7 cells. Additionally, it decreased the expression of LPS-induced p-IKKβ and p-NF-κB p65 and prevented the migration of p-NF-κB into the nucleus caused by LPS. Notably, p-JAK1, p-STAT3, Wnt 3α, β-catenin, and p-GSK-3β protein expressions were also inhibited. Therefore, the prominent anti-inflammatory activity of the P. thunbergii extract may be via the MAPK, NF-κB, JAK/STAT, Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Bin Ha
- Department of Medical Science, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Ji-Hye Kang
- Department of Medical Biotechnology, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Do Hyun Kim
- Department of Research and Development, Eshel Biopharm Co., Ltd., Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Mi-Young Lee
- Department of Medical Science, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
- Department of Medical Biotechnology, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
- Department of Research and Development, Eshel Biopharm Co., Ltd., Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| |
Collapse
|
40
|
Zhao M, Peng N, Zhou Y, Qu Y, Cao M, Zou Q, Yu Q, Lu L, Xiao F. The immunoregulatory effects of total glucosides of peony in autoimmune diseases. J Leukoc Biol 2025; 117:qiae095. [PMID: 38626175 DOI: 10.1093/jleuko/qiae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/09/2024] [Accepted: 04/08/2024] [Indexed: 04/18/2024] Open
Abstract
Total glucoside of peony and its main active ingredient paeoniflorin, extracted from the Chinese herb Paeonia lactiflora Pallas, exhibit potent immunomodulatory effects. Total glucoside of peony has been shown to inhibit inflammatory responses and disease progression in experimental models of multiple autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, Sjögren's syndrome, psoriasis, and so on. Total glucoside of peony shows broad immunomodulatory effects on many immune cells, such as T cells, macrophages, and dendritic cells, by regulating their activation, proliferation, differentiation, and production of effector molecules. Mechanistically, total glucoside of peony modulates intracellular signaling transductions, including JAK/STAT, NF-κB, MAPK, and PI3K/AKT/mTOR pathways. Moreover, total glucoside of peony has been applied in the clinical treatment of various autoimmune diseases with satisfactory therapeutic outcomes and minor side effects. Thus, available studies have demonstrated that total glucoside of peony and its bioactive constituents exhibit anti-inflammatory and immunomodulatory functions and may have extensive applications in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Mengna Zhao
- Department of Pathology, Faculty of Medicine and HKU Shenzhen Hospital, The University of Hong Kong, Pokfulam Road, 999077 Hong Kong, China
| | - Na Peng
- Department of Rheumatology, the Second People's Hospital, China Three Gorges University, 443002 Yichang, China
| | - Yingbo Zhou
- Department of Pathology, Faculty of Medicine and HKU Shenzhen Hospital, The University of Hong Kong, Pokfulam Road, 999077 Hong Kong, China
| | - Yuan Qu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, 510280 Guangzhou, China
| | - Meng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023 Jiangsu, China
| | - Qinghua Zou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, 400038 Chongqing, China
| | - Qinghong Yu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, 510280 Guangzhou, China
| | - Liwei Lu
- Department of Pathology, Faculty of Medicine and HKU Shenzhen Hospital, The University of Hong Kong, Pokfulam Road, 999077 Hong Kong, China
- Chongqing International Institute for Immunology, 401300 Chongqing, China
- Centre for Oncology and Immunology, Hong Kong Science Park, New Territories, 999077 Hong Kong, China
| | - Fan Xiao
- Department of Pathology, Faculty of Medicine and HKU Shenzhen Hospital, The University of Hong Kong, Pokfulam Road, 999077 Hong Kong, China
- Centre for Oncology and Immunology, Hong Kong Science Park, New Territories, 999077 Hong Kong, China
| |
Collapse
|
41
|
Zhu H, Ren J, Wang X, Qin W, Xie Y. Targeting skeletal interoception: a novel mechanistic insight into intervertebral disc degeneration and pain management. J Orthop Surg Res 2025; 20:159. [PMID: 39940003 PMCID: PMC11823264 DOI: 10.1186/s13018-025-05577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
Despite being a leading cause of chronic pain and disability, the underlying mechanisms of intervertebral disc (IVD) degeneration (IVDD) remain unclear. Emerging evidence suggests that mechanosensation (the ability of the skeletal system to perceive mechanical and biochemical signals) mediated by abnormal mechanical loading plays a critical role in the regulation of IVD health. This review examines the complex interactions amongIVDs, intraosseous sensory mechanisms, and the central nervous system (CNS), with a particular focus on the roles of pathways such as PGE2/EP4, Wnt/β-catenin, and NF-κB. This review elucidates the manner in which mechanical stress and aberrant signaling disrupt the homeostasis of the nucleus pulposus (NP), cartilaginous endplate (CEP) and annulus fibrosus (AF), thereby driving degeneration and exacerbating pain. Furthermore, targeted therapeutic strategies, including the modulation of skeletal interoception and dynamic mechanical loading, present novel avenues for reversing IVDD progression. By integrating skeletal biology with spinal pathology, this work offers a novel perspective on the pathogenesis of IVDD and identifies promising strategies for clinical intervention. These findings highlight the potential of targeting skeletal interoception to mitigate IVDD and associated pain, paving the way for innovative, mechanism-driven therapies.
Collapse
Affiliation(s)
- Houcheng Zhu
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - JianHang Ren
- Affiliated Yongchuan Hospital of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 402160, China
| | - Xiangjin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Wenjing Qin
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Yong Xie
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China.
| |
Collapse
|
42
|
Yamamura R, Kinoshita M, Yasumizu Y, Yata T, Kihara K, Motooka D, Shiraishi N, Sugiyama Y, Beppu S, Murata H, Koizumi N, Sano I, Koda T, Okuno T, Mochizuki H. Transcriptome signature in the blood of neuromyelitis optica spectrum disorder under steroid tapering. Front Immunol 2025; 16:1508977. [PMID: 39963140 PMCID: PMC11830620 DOI: 10.3389/fimmu.2025.1508977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Background The advent of biologics has significantly transformed treatment strategies for neuromyelitis optica spectrum disorder (NMOSD). However, there are no biomarkers that predict relapses associated with steroid tapering; therefore, it is critical to identify potential indicators of disease activity. In this study, we collected peripheral blood mononuclear cells (PBMCs) from NMOSD patients during steroid tapering and performed bulk RNA sequencing to analyze changes in immune dynamics caused by steroid reduction. Methods PBMCs were collected at 3-5 timepoints from 10 NMOSD patients at our hospital (including one relapse case), and bulk RNA sequencing was performed. All patients were positive for anti-AQP4 antibodies and had no history of biologic use. Results In one relapsed patient, gene groups with decreased expression at relapse were observed predominantly in monocytes, with upregulation in anti-inflammatory pathways such as IL-10, while the upregulated genes were related to interferon signaling. Moreover, after steroid tapering, in non-relapsed patients, genes with increased expression were enriched in inflammatory pathways, represented by interferon signaling, while genes with decreased expression were enriched in pathways related to IL-10 and glucocorticoid receptors. Weighted gene co-expression network analysis identified modules that correlated with steroid dosage, and the modules inversely correlated with steroid dosage were enriched in monocytes, with marked immune signature of interferon pathway. Conclusion This study identified peripheral blood transcriptome signatures that could lead to the identification of clinically relevant NMOSD disease activity biomarkers, and further highlights the pivotal role of interferon and IL-10 signaling in NMOSD.
Collapse
Affiliation(s)
- Ryohei Yamamura
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Makoto Kinoshita
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yoshiaki Yasumizu
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Tomohiro Yata
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Keigo Kihara
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Daisuke Motooka
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Naoyuki Shiraishi
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasuko Sugiyama
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shohei Beppu
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hisashi Murata
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naoshi Koizumi
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Itsuki Sano
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Toru Koda
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
43
|
Wei S, Li Y, Zhou J, Xia Y. Exploring MAP3K genes in gastric cancer: biomarkers, tumor microenvironment dynamics, and chemotherapy resistance. Hereditas 2025; 162:15. [PMID: 39901302 PMCID: PMC11789369 DOI: 10.1186/s41065-025-00364-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/06/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) presents a significant global health burden, necessitating a deeper understanding of its molecular underpinnings for improved diagnostics and therapeutics. METHODS In this study, we investigated the expression profiles and clinical implications of MAP3K genes in GC using in silico and in vitro experiments. RESULTS Utilizing RT-qPCR analysis, we observed significant up-regulation of MAP3K1, MAP3K4, MAP3K5, MAP3K6, MAP3K7, MAP3K8, MAP3K9, and MAP3K10 in GC cell lines, while MAP3K2, MAP3K3, MAP3K11, MAP3K12, MAP3K13, MAP3K14, and MAP3K15 exhibited down-regulation. Prognostic evaluation revealed that elevated expression of MAP3K1, MAP3K4, MAP3K7, MAP3K8, MAP3K9, and MAP3K10 was associated with shorter overall survival (OS), emphasizing their clinical significance. Furthermore, the diagnostic potential was demonstrated through robust Receiver operating characteristics (ROC) curve analysis, indicating the strong discriminatory power of these genes in distinguishing GC patients. Proteomic analysis further confirmed the higher expression of MAP3K1, MAP3K4, MAP3K7, MAP3K8, MAP3K9, and MAP3K10 genes in GC. Methylation profiling further supported the idea that promoter hypomethylation of MAP3K1, MAP3K4, MAP3K7, MAP3K8, MAP3K9, and MAP3K10 genes was associated with their up-regulation. Single-cell functional analysis elucidated the involvement of MAP3K genes in shaping the tumor microenvironment. miRNA-mRNA network analysis revealed intricate regulatory mechanisms, with hsa-mir-200b-3p emerging as a key regulator. Finally, the MAP3K1 knockdown has shown significant impacts on the cellular behavior of the BGC823 cells. CONCLUSION This comprehensive assessment provides valuable insights into the role of MAP3K genes in GC, offering avenues for further research and therapeutic exploration.
Collapse
Affiliation(s)
- Senhui Wei
- Department of Gastroenterolog, Shenzhen Guangming District People's Hospital, Shenzhen City, 518107, P.R. China
| | - Ying Li
- Department of Gastroenterolog, Shenzhen Guangming District People's Hospital, Shenzhen City, 518107, P.R. China
| | - Jing Zhou
- Department of Gastroenterolog, Shenzhen Guangming District People's Hospital, Shenzhen City, 518107, P.R. China
| | - Yongming Xia
- Department of Hepatobiliary Gastrointestinal Surgery, Shenzhen Guangming District People's Hospital, Shenzhen City, 518107, P. R. China.
| |
Collapse
|
44
|
Wang F, Wen H, Liu L, Aisa HA, Xin X. A Pair of Epimers of Lignan Alleviate Neuroinflammatory Effects by Modulating iNOS/COX-2 and MAPK/NF-κB Signaling Pathways. Inflammation 2025; 48:361-371. [PMID: 38878150 DOI: 10.1007/s10753-024-02080-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 02/09/2025]
Abstract
Neuroinflammation is a causative factor in neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis. Previous studies have shown that Artemisia mongolica has anti-inflammatory properties. Aschantin (AM3) has been shown to have anti-inflammatory effects. However, the mechanism of AM3 and its epimer epi-aschantin (AM2) remains controversial. Therefore, the present study explored the mechanism of neuroinflammation by AM2 and AM3 and attempted to reveal the relationship between the structure of AM2 and AM3 and anti-neuroinflammatory activity. We isolated for the first time 12 lignans from A. mongolica that inhibited NO content at 10 μM in LPS-stimulated BV2 cells. Among them, epi-aschantin (AM2) and Aschantin (AM3) showed significant inhibition in NO screening. With further studies, we found that both AM2 and AM3 effectively inhibited the overproduction of NO, PGE2, IL-6, TNF-α and MCP-1, as well as the overexpression of COX-2 and iNOS. Mechanistic studies have shown AM2 and AM3 significantly inhibited the phosphorylation of ERK, JNK and P-38 in the MAPK signaling pathway and p-IκBα,p-p65 and blocked p65 entry into the nucleus. The results suggested that the pair of epimers (AM2 and AM3) can be used as potential therapeutic agents in the treatment of various brain disorders and that structural differences do not differ in anti-neuroinflammatory effects.
Collapse
Affiliation(s)
- Fangsheng Wang
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Huizhen Wen
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Liu Liu
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
| | - Haji Aakber Aisa
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.
| | - Xuelei Xin
- The State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.
| |
Collapse
|
45
|
Kim HS, Ahn JW, Park JY, Joo SS. Identification, characterization, and anti-inflammatory activity of a lipocalin-like protein cloned from Oenanthe javanica. Food Sci Biotechnol 2025; 34:721-731. [PMID: 39958172 PMCID: PMC11822164 DOI: 10.1007/s10068-024-01700-2] [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: 03/26/2024] [Revised: 08/08/2024] [Accepted: 08/23/2024] [Indexed: 02/18/2025] Open
Abstract
This study investigated the anti-inflammatory effects of OJlipo1, a lipocalin-like protein derived from Oenanthe javanica. Through cloning and expressing OJlipo1 in E. coli, and subsequent rigorous characterization including amino acid analysis and mass spectrometry, its potential against inflammation was evaluated. Studies on lipopolysaccharide-stimulated RAW 264.7 cells highlighted its capability to suppress nitric oxide synthase and cyclooxygenase-2 expression, as well as its interference with nuclear factor kappa B and mitogen-activated protein kinase pathways, which are essential for toll-like receptor 4 (TLR4) signaling. Utilizing TAK242, a TLR4 pathway inhibitor, reinforced OJlipo1's specific targeting mechanism. These findings underscore OJlipo1's significant anti-inflammatory potential, aligning with the traditional uses of O. javanica, and suggest new therapeutic avenues, especially for diseases associated with TLR4 dysregulation. This validates the traditional application of O. javanica in inflammation and positions OJlipo1 as a promising therapeutic candidate, enriching our understanding of its molecular underpinnings and therapeutic prospects.
Collapse
Affiliation(s)
- Hyun Soo Kim
- Department of Marine Life Science, College of Life Science, Gangneung-Wonju National University, Gangneung, 25457 Gangwon Republic of Korea
| | - Jeong Won Ahn
- Department of Marine Life Science, College of Life Science, Gangneung-Wonju National University, Gangneung, 25457 Gangwon Republic of Korea
| | - Jung Youl Park
- Glocal University Project Group, Andong National University, 1375 Gyeongdong-ro, Andong, Gyeongsangbuk-do 36729 Republic of Korea
| | - Seong Soo Joo
- Department of Marine Life Science, College of Life Science, Gangneung-Wonju National University, Gangneung, 25457 Gangwon Republic of Korea
- Huscion MAJIC R&D Center, 331 Pangyo-ro, Seongnam, 13488 Gyeonggi Republic of Korea
| |
Collapse
|
46
|
Goh RCW, Maharajan MK, Gopinath D, Fang CM. The Therapeutic Effects of Probiotic on Systemic Lupus Erythematosus in Lupus Mice Models: A Systematic Review. Probiotics Antimicrob Proteins 2025; 17:35-50. [PMID: 38806966 DOI: 10.1007/s12602-024-10297-1] [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] [Accepted: 05/18/2024] [Indexed: 05/30/2024]
Abstract
Increasing evidence suggests the beneficial immunomodulatory effects of probiotics can reduce inflammation in systemic lupus erythematosus (SLE). However, there is no summary of the existing evidence available. This study aims to investigate the therapeutic effects of probiotics on SLE in a lupus mouse model by examining various markers, including inflammatory cytokines, Treg cells, disease activity, and gut microbiota. A systematic search was conducted using three databases (Web of Science, PubMed, and Scopus) to identify animal studies that reported the therapeutic benefits of probiotics against SLE. Data extracted from the selected articles were qualitatively synthesized. The SYRCLE risk of bias tool was used to evaluate the risk of bias. Out of a total of 3205 articles, 12 met the inclusion criteria. Probiotic strains, quantities, and routes of administration varied among the studies. The treatment ranged from 8 to 47 weeks. Probiotic strains such as L. fermentum CECT5716, L. casei B255, L. reuteri DSM 17509, L. plantarum LP299v, and L. acidophilus can significantly reduce pro-inflammatory cytokines (TNF-α, IL-12, IL-6, IL-1β, IL-17, and IFN-γ) levels while increasing anti-inflammatory IL-10 and Treg cells. Probiotics also delay the production of autoantibodies, thus prolonging the remission period, decreasing flare frequency, and delaying disease progression. Furthermore, probiotic administration prevents gut dysbiosis, increases intestinal stability, and prevents pathogen colonization. In conclusion, probiotics can be considered a new alternative therapeutic approach for the management of SLE. Further clinical studies are required to investigate and validate the safety and effectiveness of probiotics in humans.
Collapse
Affiliation(s)
- Rachael Chaeh-Wen Goh
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia
| | - Mari Kannan Maharajan
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia
| | - Divya Gopinath
- Basic Medical and Dental Sciences Department, College of Dentistry, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Malaysia.
| |
Collapse
|
47
|
Zhao H, Mu Y, Liang A, Wei J, Lai S, Li X, Chen P, Li H, He H, Liu X, Liu H. Suppressing DUSP16 overexpression induced by ELK1 promotes neural progenitor cell differentiation in mouse models of Alzheimer's disease. Aging Cell 2025; 24:e14372. [PMID: 39434411 PMCID: PMC11822628 DOI: 10.1111/acel.14372] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Emerged evidence indicated that stimulating hippocampal neurogenesis is a potential strategy for restoring cognition in AD. Mitogen-activated protein kinases (MAPKs) play an essential role in neurogenesis. Meanwhile, the enzymatic power of the phosphatases is much greater than that of kinases. Dual-specificity phosphatase 16 (DUSP16), known to as a phosphatase negatively regulate MAPKs, may be implicated in neural differentiation. Nevertheless, the effect of DUSP16 on cognitive disorders by stimulating neural progenitor cell (NPC) differentiation in AD mice remains unclear. Our study demonstrates an association between DUSP16 SNPs and clinical progression in individuals with mild cognitive impairment (MCI). Besides, increased DUSP16 expression was detected in both 3xTg and SAMP8 mouse models of AD, accompanied by NPC neural differentiation impairments. By silencing DUSP16, the induction of neural differentiation, synaptic transmission, and cognitive benefits were observed in both AD mice. Furthermore, DUSP16 was involved in the process of NPC differentiation through regulating c-Jun N-terminal kinase (JNK) phosphorylation and SOX2 expression. Moreover, ETS transcription factor (ELK1) was involved in the DUSP16 transcription, which resulted in the upregulation of DUSP16 at the state of AD. Our data uncovers a potential regulatory role for DUSP16 in adult hippocampal neurogenesis (AHN) and provides a possibility to find a novel strategy for AD intervention.
Collapse
Affiliation(s)
- Huimin Zhao
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Yao Mu
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Anqi Liang
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Jie Wei
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Sixian Lai
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Xin Li
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Peipei Chen
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Hao Li
- Acupuncture and Moxibustion DepartmentJiangsu Provincial Second Chinese Medicine Hospital/the Second Affiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
| | - Hua He
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Xiaoquan Liu
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Haochen Liu
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
48
|
Witowski A, Palmowski L, Rahmel T, Nowak H, Ehrentraut SF, Putensen C, von Groote T, Zarbock A, Babel N, Anft M, Sitek B, Bracht T, Bayer M, Weber M, Weisheit C, Pfänder S, Eisenacher M, Adamzik M, Katharina R, Koos B, Ziehe D. Activation of the MAPK network provides a survival advantage during the course of COVID-19-induced sepsis: a real-world evidence analysis of a multicenter COVID-19 Sepsis Cohort. Infection 2025; 53:107-115. [PMID: 38896372 PMCID: PMC11825614 DOI: 10.1007/s15010-024-02325-7] [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: 02/13/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE There is evidence that lower activity of the RAF/MEK/ERK network is associated with positive outcomes in mild and moderate courses of COVID-19. The effect of this cascade in COVID-19 sepsis is still undetermined. Therefore, we tested the hypothesis that activity of the RAF/MEK/ERK network in COVID-19-induced sepsis is associated with an impact on 30-day survival. METHODS We used biomaterial from 81 prospectively recruited patients from the multicentric CovidDataNet.NRW-study cohort (German clinical trial registry: DRKS00026184) with their collected medical history, vital signs, laboratory parameters, microbiological findings and patient outcome. ERK activity was measured by evaluating ERK phosphorylation using a Proximity Ligation Assay. RESULTS An increased ERK activity at 4 days after diagnosis of COVID-19-induced sepsis was associated with a more than threefold increased chance of survival in an adjusted Cox regression model. ERK activity was independent of other confounders such as Charlson Comorbidity Index or SOFA score (HR 0.28, 95% CI 0.10-0.84, p = 0.02). CONCLUSION High activity of the RAF/MEK/ERK network during the course of COVID-19 sepsis is a protective factor and may indicate recovery of the immune system. Further studies are needed to confirm these results.
Collapse
Affiliation(s)
- Andrea Witowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Lars Palmowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Tim Rahmel
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Hartmuth Nowak
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
- Zentrum für Künstliche Intelligenz, Medizininformatik und Datenwissenschaften, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Stefan F Ehrentraut
- Klinik für Anästhesiologie und operative Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Christian Putensen
- Klinik für Anästhesiologie und operative Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Thilo von Groote
- Klinik für Anästhesiologie, operative Intensivmedizin und Schmerztherapie, Universitätsklinikum Münster, Münster, Germany
| | - Alexander Zarbock
- Klinik für Anästhesiologie, operative Intensivmedizin und Schmerztherapie, Universitätsklinikum Münster, Münster, Germany
| | - Nina Babel
- Centrum für Translationale Medizin, Medizinische Klinik I, Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, Herne, Germany
| | - Moritz Anft
- Centrum für Translationale Medizin, Medizinische Klinik I, Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, Herne, Germany
| | - Barbara Sitek
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Medizinische Fakultät, Bochum, Germany
| | - Thilo Bracht
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Medizinische Fakultät, Bochum, Germany
| | - Malte Bayer
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Medizinische Fakultät, Bochum, Germany
| | - Maike Weber
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Medizinische Fakultät, Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr Universität Bochum, Bochum, Germany
| | - Christina Weisheit
- Klinik für Anästhesiologie und operative Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Stephanie Pfänder
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- University of Lübeck, Lübeck, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Medizinische Fakultät, Bochum, Germany
- Center for Protein Diagnostics (PRODI), Medical Proteome Analysis, Ruhr Universität Bochum, Bochum, Germany
| | - Michael Adamzik
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Rump Katharina
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Björn Koos
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany.
| | - Dominik Ziehe
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| |
Collapse
|
49
|
Gu X, Chen C, Chen Y, Zeng C, Lin Y, Guo R, Xu S, Lin C. Bioinformatics approach reveals the critical role of inflammation-related genes in age-related hearing loss. Sci Rep 2025; 15:2687. [PMID: 39837906 PMCID: PMC11751394 DOI: 10.1038/s41598-024-83428-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] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory impairment in the elderly. However, the pathogenesis of ARHL remains unclear. This study was aimed to explore the potential inflammation-related genes of ARHL and suggest novel therapeutic targets for this condition. Initially, a total of 105 Inflammatory related differentially expressed genes (IRDEGs) were obtained by overlapping the differentially expressed genes from the GSE49522 and GSE49543 datasets with Inflammatory related genes. The IRDEGs were mainly enriched in MAPK, PI3K-Akt, Hippo and JAK-STAT pathways by analysis of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. We then identified 10 key IRDEGs including Alox5ap, Chil1, Clec7a, Dysf, Fcgr3, etc. using Least absolute shrinkage and selection operator regression analysis and converted them into human genes. The ROC curve indicated that Alox5ap expression presented a high accuracy in distinguishing between different groups. By CIBERSORT algorithm, 8 humanized key IRDEGs were correlated with the infiltration abundance of 3 immune cells. Finally, it showed that the Alox5ap expression was significantly more effective compared to other variables in the diagnostic model of ARHL. This study suggests that inflammation might play a role in the development of ARHL, providing a deeper understanding of the underlying causes of this disease.
Collapse
Affiliation(s)
- Xi Gu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chenyu Chen
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Yuqing Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chaojun Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yanchun Lin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruosi Guo
- Fujian Medical University, Fuzhou, China
| | - Shujin Xu
- Fujian Medical University, Fuzhou, China
| | - Chang Lin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
50
|
Chen C, He J, Huang W, Xu D, Li Z, Yang A. PLK3 weakens antioxidant defense and inhibits proliferation of porcine Leydig cells under oxidative stress. Sci Rep 2025; 15:2612. [PMID: 39837970 PMCID: PMC11751325 DOI: 10.1038/s41598-025-86867-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025] Open
Abstract
Aging is characterized by cellular degeneration and impaired physiological functions, leading to a decline in male sexual desire and reproductive capacity. Oxidative stress (OS) lead to testicular aging by impairing the male reproductive system, but the potential mechanisms remain unclear. In the present study, the functional status of testicular tissues from young and aged boars was compared, and the transcriptional responses of Leydig cells (LCs) to hydrogen peroxide (H2O2)-induced senescence were explored, revealing the role of OS in promoting aging of the male reproductive system. 601 differentially expressed genes (DEGs) associated with OS, cell cycle regulation, and intracellular processes were identified. These DEGs were significantly enriched in critical aging pathways, including the p53 signaling pathway, autophagy, and cellular senescence. Protein-protein interaction (PPI) network analysis unveiled 15 key genes related to cell cycle and DNA replication, with polo-like kinase 3 (PLK3) exhibiting increased expression under OS. In vitro, PLK3 knockdown significantly enhanced the viability and antioxidant capacity of LCs under OS. This study deepens our understanding of how LCs respond to OS and provides new therapeutic targets for enhancing cellular resistance to oxidative damage and promoting tissue health.
Collapse
Affiliation(s)
- Chujie Chen
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- College of Life Sciences and Resource Environment, Yichun university, Yichun, Jiangxi, China
| | - Jinyan He
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weixian Huang
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Dong Xu
- Department of Biological and Environmental Engineering, Yueyang Vocational Technical College, Yueyang, Hunan, China
| | - Zhaohui Li
- Xiangtan Livestock Breeding Station, Xiangtan, Hunan, China
| | - Anqi Yang
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|