1
|
Lin J, Ma J, Wang Z, Xu C, Sun Y, Miao Z, Chen Z, Sun Z, Zhang X, Wu Y. DBC1 promotes intervertebral disc degeneration by activating NF-κB pathway and inhibiting SIRT1 activity. Life Sci 2025; 373:123689. [PMID: 40339956 DOI: 10.1016/j.lfs.2025.123689] [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: 01/20/2025] [Revised: 04/21/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
AIMS Intervertebral disc degeneration (IVDD) is a leading contributor to spinal degenerative diseases; however, its pathogenesis remains only partially elucidated. Recent studies have highlighted that the diminished activity of SIRT1 and the aberrant activation of the NF-κB signaling pathway are critical pathogenic factors in IVDD. DBC1 has been identified as a regulator of SIRT1 activity and the NF-κB signaling pathway. This study aimed to investigate the role of DBC1 in IVDD. MATERIALS AND METHODS The expression levels of DBC1 in the nucleus pulposus of aging rats were quantified. Both overexpression and knockdown of DBC1 were utilized to explore their effects on the extracellular matrix (ECM) of the nucleus pulposus. Furthermore, the influence of DBC1 on cellular senescence, apoptosis, and ECM regulation in nucleus pulposus cells was assessed using Western blot (WB), cellular fluorescence assays, and histological staining techniques. KEY FINDINGS Our results demonstrate that DBC1 expression is significantly upregulated in IVDD. Moreover, DBC1 appears to contribute to IVDD by promoting apoptosis, senescence, and ECM degradation in nucleus pulposus cells. Mechanistic investigations revealed that DBC1 activates the NF-κB signaling pathway while suppressing SIRT1 expression in nucleus pulposus cells, suggesting that these two mechanisms underlie its effects on IVDD. SIGNIFICANCE In summary, this study provides evidence that DBC1 may play a pivotal role in the pathogenesis of IVDD by inhibiting SIRT1 activity and activating the NF-κB signaling pathway. Consequently, targeting DBC1 suppression could represent a promising therapeutic strategy for managing IVDD.
Collapse
Affiliation(s)
- Jiahao Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiawei Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ze Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Cong Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yun Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhimin Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zexin Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zeming Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Yaosen Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
2
|
Shawky A, Saber S, Abd El-Kader EM, El-Kashef HA. Verapamil inhibits TXNIP-dependent NLRP3 Inflammasome activation in an ulcerative colitis rat model: A new evolving role of the calcium channel blocker. Int Immunopharmacol 2025; 158:114751. [PMID: 40359884 DOI: 10.1016/j.intimp.2025.114751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Ulcerative colitis (UC) is a long-term inflammatory bowel disease (IBD) associated with significant morbidity. It is marked by inflammation and damage to the colon's mucosal lining. Studies have shown that NLRP3 inflammasome activation, apoptosis, and impaired autophagy are critical in its pathogenesis. Verapamil, a calcium channel blocker, has been found to inhibit NLRP3 inflammasome activation in various preclinical models. However, the potential influence of verapamil on the TXNIP in UC remains unexplored. This study investigates the effects of verapamil on an UC rat model induced chemically by acetic acid. Verapamil effectively inhibited the TXNIP-NLRP3-caspase-1 axis, reducing inflammasome activation and the release of IL-1β and IL-18. Additionally, verapamil suppressed NFκB, the priming step of NLRP3 activation. The drug enhanced autophagic activity, as indicated by increased expression of LC3-II and Beclin-1, along with reduced LC3-I and mTOR expression. Moreover, it demonstrated anti-apoptotic effects mediated by regulating Bax and cleaved caspase-3. These molecular changes contributed to mucosal healing and improved microscopic and macroscopic outcomes in the colitis model. Furthermore, verapamil improved the colon weight-to-length ratio and disease activity scores and mitigated oxidative stress. As verapamil has been safely used in clinics to treat hypertension, our findings suggest it may be a safe therapeutic option for ameliorating inflammation and apoptosis and activating autophagy in UC pathology. Since hypertension demonstrates a strong association with UC, the use of verapamil merits particular attention in hypertensive patients fighting against IBD.
Collapse
Affiliation(s)
- Ahmed Shawky
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Eman M Abd El-Kader
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Hassan A El-Kashef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
3
|
Liao H, Zheng J, Lu J, Shen HL. NF-κB Signaling Pathway in Rheumatoid Arthritis: Mechanisms and Therapeutic Potential. Mol Neurobiol 2025; 62:6998-7021. [PMID: 39560902 DOI: 10.1007/s12035-024-04634-2] [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: 03/11/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that imposes a heavy economic burden on patients and society. Bone and cartilage destruction is considered an important factor leading to RA, and inflammation, oxidative stress, and mitochondrial dysfunction are closely related to bone erosion and cartilage destruction in RA. Currently, there are limitations in the clinical treatment methods for RA, which urgently necessitates finding new effective treatments for patients. Nuclear transcription factor-κB (NF-κB) is a signaling transcription factor that is widely present in various cells. It plays an important role as a stress source in the cellular environment and regulates gene expression in processes such as immunity, inflammation, cell proliferation, and apoptosis. NF-κB has long been recognized as a pathogenic factor of RA, and its activation can exacerbate RA by promoting inflammation, oxidative stress, mitochondrial dysfunction, and bone destruction. Conversely, inhibiting the activity of the NF-κB pathway effectively inhibits these pathological processes, thereby alleviating RA. Therefore, NF-κB may be a potential therapeutic target for RA. This article describes the physiological structure of NF-κB and its important role in RA through the regulation of oxidative stress, inflammatory response, mitochondrial function, and bone destruction. Meanwhile, we also summarized the impact of NF-κB crosstalk with other signaling pathways on RA and the effect of related drugs or inhibitors targeting NF-κB on RA. The purpose of this article is to provide evidence for the role of NF-κB in RA and to emphasize its significant role in RA by elucidating the mechanisms, so as to provide a theoretical basis for targeting the NF-κB pathway as a treatment for RA.
Collapse
Affiliation(s)
- Haiyang Liao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jianxiong Zheng
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jinyue Lu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Hai-Li Shen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
4
|
Saamarthy K, Daams R, Sime W, Persson C, Chygorin E, Ahlqvist K, Evans-Axelsson S, Strand D, Massoumi R. An optimised Bcl-3 inhibitor for melanoma treatment. Br J Pharmacol 2025; 182:2426-2446. [PMID: 39943627 DOI: 10.1111/bph.17467] [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: 09/04/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Malignant melanoma is the most lethal form of skin cancer, characterised by a poor survival rate. One of the key factors driving the aggressive growth of melanoma cells is the elevated expression of the proto-oncogene Bcl-3. This study aims to optimise, evaluate and characterise a second-generation Bcl-3 inhibitor, using melanoma as a model to demonstrate its potential therapeutic efficacy. EXPERIMENTAL APPROACH We synthesised and screened a series of structural analogues and selected A27, the most promising candidate for further investigation. We assessed whether A27 disrupted the interaction between Bcl-3 and its binding partner, p50, and examined the subsequent effects on cyclin D1 expression. Additionally, we evaluated the impact of A27 on melanoma cell proliferation and migration in vitro, as well as its therapeutic efficacy in various in vivo melanoma models. KEY RESULTS Nuclear magnetic resonance (NMR) confirmed that A27 directly binds to Bcl-3, effectively inhibiting its function. By disrupting the Bcl-3/p50 interaction, A27 led to a significant down-regulation of cyclin D1 expression. In cellular assays, A27 markedly reduced proliferation and migration of melanoma cells. In vivo, treatment with A27 resulted in a substantial reduction in melanoma tumour growth, with no observed toxicity in treated animals. CONCLUSIONS AND IMPLICATIONS At present, no other Bcl-3 inhibitors exist for clinical application in the field of oncology, and as a result, our novel findings provide a unique opportunity to develop a highly specific drug against malignant melanoma to meet an urgent clinical need.
Collapse
Affiliation(s)
- Karunakar Saamarthy
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Cecilia Persson
- Swedish NMR Center, Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Eduard Chygorin
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Kristofer Ahlqvist
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Susan Evans-Axelsson
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| | - Daniel Strand
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Huang JC, Tong XL, Xiang MSW, Boumelhem BB, Foulis DP, Zhang M, McKenzie CA, McCaughan GW, Reinheckel T, Zhang HE, Gorrell MD. Dipeptidyl peptidase 9 (DPP9) depletion from hepatocytes in experimental primary liver cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167819. [PMID: 40187163 DOI: 10.1016/j.bbadis.2025.167819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Dipeptidyl peptidase 9 (DPP9) is an indispensable intracellular protease. Among its many molecular functions is suppression of the NLRP1 inflammasome. Inhibitors targeting all four proteases of the DPP4 family, including DPP9, can reduce tumour burden, including in mouse liver. To explore hepatocyte DPP9 in experimental hepatocellular carcinoma (HCC), we generated hepatocyte-specific DPP9-KO mice by crossing albumin-Cre mice with DPP9 floxed mice and treated sequentially with diethylnitrosamine, then with thioacetamide combined with an atherogenic high-fat diet until 28 weeks of age. DPP9-KO mice had less body, liver and subcutaneous adipose tissue mass, lower fasting plasma glucose and fewer small macroscopic liver nodules compared to DPP9-WT control mice. However, there were no differences in the total number of macroscopic liver nodules, or of microscopic tumour burden, inflammation, fibrosis or steatosis. Consistent with the known function of DPP9 to suppress NLRP1 activation, activated caspase-1 protein and inflammation markers Nfkbib, Cxcl10 and Ccl5 were elevated in DPP9-KO liver. The tumour suppressor protein p53 was increased and the autophagy proteins beclin1, LC3B and p62 were altered. In conclusion, hepatocyte-specific DPP9 gene deletion in experimental primary liver cancer improved energy metabolism and may reduce liver cancer initiation, via mechanisms that may include increased autophagy and tumour suppression.
Collapse
MESH Headings
- Animals
- Hepatocytes/pathology
- Hepatocytes/metabolism
- Hepatocytes/enzymology
- Mice
- Mice, Knockout
- Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics
- Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism
- Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/deficiency
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/chemically induced
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Male
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Diet, High-Fat/adverse effects
- Mice, Inbred C57BL
- Inflammasomes/metabolism
- Liver/pathology
- Liver/metabolism
Collapse
Affiliation(s)
- JiaLi Carrie Huang
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Xinlin Linda Tong
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michelle Sui Wen Xiang
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Badwi B Boumelhem
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Diarmid P Foulis
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - MingChang Zhang
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Catriona A McKenzie
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Geoffrey W McCaughan
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, Australia
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Hui E Zhang
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Mark D Gorrell
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
6
|
Lai X, Zhang X, Lai J, Zhao W, Song Z, Chen Y, Ud din M, Munawer MF, Jiang H, Liu X, Wang X. Targeted self-assembled anti-NFκB AuNCs-aptamer nanoplatform for precise theranostics via tailored follicle regeneration. Mater Today Bio 2025; 32:101774. [PMID: 40290889 PMCID: PMC12032944 DOI: 10.1016/j.mtbio.2025.101774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
NFκB is a vital transcription factor for the regulation of hair follicle cycle. As a therapeutic target, NFκB is specifically blocked by RNA aptamer with negligible side effects, but the targeted transmembrane transport of anti-NFκB aptamer remains a challenge due to its negative charge under physiological conditions. In this study, taking advantage of the depilation-induced oxidative stress microenvironment (OSM), it was confirmed for the first time that self-assembled gold nanoclusters and aptamer (AuNCs-Aptamer) complexes formed in the skin and enhanced the therapeutic effect of anti-NFκB aptamer drugs, effectively blocking the NFκB-mediated inflammatory response and inhibiting hair follicle regeneration. The hematoxylin-eosin (HE) staining of tissue section and hematology analysis demonstrated that OSM-responsive self-assembled AuNCs-Aptamer caused no toxicity to the living organism. Moreover, self-assembly occurred only in the oxidative stress-injured skin cells rather than the normal cells, which revealed that this self-assembly was a targeted, safe and effective therapy for hypertrichosis.
Collapse
Affiliation(s)
- Xiangdong Lai
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiaoyang Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jiejuan Lai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weiwei Zhao
- State Key Laboratory Breeding Base for The Protection and Utilization of Biological Resources in Tarim Basin, College of Life Science and Technology, Tarim University, Alar, Xinjiang, 843300, China
| | - Zhongquan Song
- Department of Pulmonary and Critical Care Medicine, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Yuanyuan Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Miraj Ud din
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Muhammad Faizan Munawer
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
7
|
Sadeghi T, Zaki-Dizaji M, Dolatabad MR, Taheri Z. MiR-568-5p, the key regulator of suppressor of cytokine signaling 4 (SOCS4) in the coronary artery disease. J Diabetes Metab Disord 2025; 24:106. [PMID: 40276596 PMCID: PMC12014962 DOI: 10.1007/s40200-025-01619-3] [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: 12/25/2024] [Accepted: 04/06/2025] [Indexed: 04/26/2025]
Abstract
Objectives Pro-inflammatory molecules are key risk factors for coronary artery diseases (CAD). Therefore, the regulation of inflammatory responses plays a crucial role in CAD. Suppressor of cytokine signaling 4 (SOCS4) is a negative regulator of cytokine signaling may significantly contribute to the pathogenesis of CAD. Bioinformatics studies indicate that hsa-miR-568-5p can target SOCS4. This study aimed to evaluate the expression of hsa-miR-568-5p in patients with CAD and investigate its correlation with SOCS4 levels. Methods The study included 20 Iranian participants without artery stenosis and 40 with artery stenosis. The relative expression levels of hsa-miR-568-5p and SOCS4 were assessed using Real-Time PCR. Results The findings revealed no significant difference in the expression levels of hsa-miR-568-5p and SOCS4 between the two groups. Also, correlations were not observed between hsa-miR-568-5p, SOCS4, and age in both the control and CAD patient groups. Conclusions While hsa-miR-568-5p regulates SOCS4 expression, and SOCS4 upregulation is implicated in coronary artery disease (CAD) protection, this study found no conclusive evidence supporting this relationship. Further research is warranted.
Collapse
Affiliation(s)
- Tayebeh Sadeghi
- Department of Physiology, Ke.C., Islamic Azad University, Kerman, Iran
| | - Majid Zaki-Dizaji
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Zahra Taheri
- Department of Biology, SR. C., Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
Jayab NA, Abed A, Talaat IM, Hamoudi R. The molecular mechanism of NF-κB dysregulation across different subtypes of renal cell carcinoma. J Adv Res 2025; 72:501-514. [PMID: 39094893 DOI: 10.1016/j.jare.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The nuclear factor kappa B (NF-κB) is a critical pathway that regulates various cellular functions, including immune response, proliferation, growth, and apoptosis. Furthermore, this pathway is tightly regulated to ensure stability in the presence of immunogenic triggers or genotoxic stimuli. The lack of control of the NF-κB pathway can lead to the initiation of different diseases, mainly autoimmune diseases and cancer, including Renal cell carcinoma (RCC). RCC is the most common type of kidney cancer and is characterized by complex genetic composition and elusive molecular mechanisms. AIM OF REVIEW The current review summarizes the mechanism of NF-κB dysregulation in different subtypes of RCC and its impact on pathogenesis. KEY SCIENTIFIC CONCEPT OF REVIEW This review highlights the prominent role of NF-κB in RCC development and progression by driving the expression of multiple genes and interplaying with different pathways, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. In silico analysis of RCC cohorts and molecular studies have revealed that multiple NF-κB members and target genes are dysregulated. The dysregulation includes receptors such as TLR2, signal-transmitting members including RelA, and target genes, for instance, HIF-1α. The lack of effective regulatory mechanisms results in a constitutively active NF-κB pathway, which promotes cancer growth, migration, and survival. In this review, we comprehensively summarize the role of dysregulated NF-κB-related genes in the most common subtypes of RCC, including clear cell RCC (ccRCC), chromophobe RCC (chRCC), and papillary RCC (PRCC).
Collapse
Affiliation(s)
- Nour Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Alaa Abed
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Iman M Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, 21131 Alexandria, Egypt.
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, 27272 Sharjah, United Arab Emirates; Division of Surgery and Interventional Science, University College London, London, United Kingdom; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
9
|
Silva Porta C, de Souza AP, Conter FU, Picada JN, da Silva J, Yunes JS, Marinowic DR, Dihl RR. Integrative assessment of the genotoxic effects of the neurotoxin saxitoxin produced by the freshwater cyanobacterium Raphidiopsis raciborskii. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025:1-12. [PMID: 40401712 DOI: 10.1080/15287394.2025.2509761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Saxitoxin (STX), a potent neurotoxin produced by cyanobacteria, has not been comprehensively investigated with respect to genotoxic potential, especially in freshwater environments. This study aimed to characterize the genotoxic potential of STX obtained from Raphidiopsis. raciborskii cultures using in vitro and in silico approaches. Mutagenic potential was determined through the Ames test with Salmonella typhimurium strains TA98, TA100, and TA102. DNA damage and chromosomal instability were assessed in human glioblastoma U87-MG cells using the comet and cytokinesis-block micronucleus cytome (CBMN-Cyt) assay, respectively. In addition, systems biology tools were applied to explore STX interactions with genes involved in DNA damage response pathways. Data demonstrated no marked mutagenic activity in the Ames test across tested concentrations (0.625-10 µg/L). However, significant DNA damage and increased micronucleus (MN) formation were observed at 2.5, 5, or 10 µg/L in U87-MG cells, without accompanying cytotoxicity. In silico analysis identified interactions between STX and key proteins, including P53, CDK5, and GSK3B, indicating pathways related to DNA damage, cell cycle regulation, and neurogenesis. These findings suggest that STX from freshwater cyanobacteria might induce genotoxic effects at environmentally relevant concentrations. The integration of in vitro and computational data supports the need for regulatory monitoring of STX in drinking water and emphasizes the relevance of neural cell-based models in assessing cyanotoxin-related adverse risks.
Collapse
Affiliation(s)
- Cynthia Silva Porta
- Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Ana Paula de Souza
- Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Felipe Umpierre Conter
- Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Jaqueline Nascimento Picada
- Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, Brazil
- Institute of Basic Health Sciences (ICBS), Department of Pharmacology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Juliana da Silva
- Laboratory of Genetics Toxicology, La Salle University, Canoas, Brazil
| | - João Sarkis Yunes
- Institute of Oceanography, Laboratory of Cyanobacteria and Phycotoxins, Federal University of Rio Grande (FURG), Rio Grande, Brazil
| | - Daniel Rodrigo Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rafael Rodrigues Dihl
- Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil (ULBRA), Canoas, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
10
|
Shamnewadi A, Unger BS, Palit P, Mallapur SP, Patil VS, Darasaguppe Ramachandra H, Ikbal AMA, Jalalpure SS. In Silico and In Vivo Pharmacological Study of Acmella paniculata Flowers for Anti-Inflammatory and Antiarthritic Potential. Chem Biodivers 2025:e00428. [PMID: 40387128 DOI: 10.1002/cbdv.202500428] [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: 02/03/2025] [Revised: 04/23/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Acmella paniculata has been traditionally used in folklore medicine to alleviate pain and manage articular rheumatism. This study explores its potential anti-inflammatory and antiarthritic effects through in silico and in vivo approaches. A. paniculata bioactives' antiarthritic mechanisms were elucidated using computational techniques, namely, gene set enrichment analysis, network pharmacology, molecular docking, and molecular dynamics (MD) simulations using KEGG pathway analysis, PyRx, Discovery Studio, and GROMACS tools. A. paniculata hydroalcoholic extract (APE) and the ethyl acetate fraction (APF) were analyzed via LC‒MS for phytochemical profiling. In vivo studies assessed anti-inflammatory and antiarthritic potential in carrageenan-induced paw edema and complete Freund's adjuvant (CFA)-induced arthritis models in Wistar rats. Ferulic acid, isoferulic acid, and acetyl aleuritolic acid were identified as bioactives that targeted RELA, a key NF-κB component. Stable interactions were confirmed through docking and MD simulations. LC‒MS verified these compounds in APE and APF. In vivo study revealed significant reductions in paw volume, arthritis scores, and inflammatory markers (CRP, RF, IL-6, and TNF-α) and improved histopathological outcomes in the APE and APF-treated groups compared to the CFA. These findings highlight the anti-inflammatory and antiarthritic potential of A. paniculata via multi-protein modulation, particularly NF-κB signaling, and it can be utilized as a promising therapeutic for rheumatoid arthritis.
Collapse
Affiliation(s)
- Akshay Shamnewadi
- ICMR-National Institute of Traditional Medicine, Belagavi, India
- KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, India
| | - Banappa S Unger
- ICMR-National Institute of Traditional Medicine, Belagavi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Partha Palit
- Department of Pharmaceutical Sciences, Assam University (A Central University), Silchar, India
| | - Shamanand P Mallapur
- ICMR-National Institute of Traditional Medicine, Belagavi, India
- KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, India
| | - Vishal S Patil
- ICMR-National Institute of Traditional Medicine, Belagavi, India
- KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, India
| | - Harish Darasaguppe Ramachandra
- ICMR-National Institute of Traditional Medicine, Belagavi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Abu Md Ashif Ikbal
- Department of Pharmaceutical Sciences, Drug Discovery Research Laboratory, Assam University, Silchar, India
| | - Sunil S Jalalpure
- KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, India
| |
Collapse
|
11
|
Andrade-Feraud CM, Acanda de la Rocha AM, Berlow NE, Duque S, Velazco A, Castillo D, Holcomb B, Coats ER, Ghurani YR, Lucey CM, Pearson B, Guilarte TR, Azzam DJ. Chronic arsenic exposure of ovarian surface and fallopian tube cultures induces giant and/or multinucleated cells with phagocytosis-like properties and an inflammatory phenotype. Toxicol Appl Pharmacol 2025; 500:117394. [PMID: 40368219 DOI: 10.1016/j.taap.2025.117394] [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: 01/31/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Chronic exposure to arsenic, a toxic metalloid frequently found in groundwater and food, represents a significant environmental health risk and has been implicated in the etiology of several cancers, including ovarian cancer. However, the precise pathways through which arsenic exerts its toxic impact on the ovary are not fully understood. This study investigates the impact of chronic arsenic exposure at environmentally relevant concentrations (75 ppb or μg/L) on primary human ovarian surface (OCE1) and fallopian tube (FNE1) cultures derived from the same donor. These heterogeneous cultures provide a unique, human-relevant platform to investigate how chronic arsenic exposure influences distinct cell types within a shared microenvironment. Prolonged arsenic exposure induced significant cytotoxicity and promoted the formation of giant and/or multinucleated cells in both cultures. These cells exhibited phagocytosis-like properties, actively engulfing apoptotic debris. Transcriptomic analyses and pathway enrichment revealed robust activation of pro-inflammatory signaling, notably the canonical NF-κB pathway. This was marked by nuclear translocation of the NF-κB p65 subunit and elevated expression and secretion of pro-inflammatory cytokines, including TNFα, IL-6, and IL-8, driving a sustained inflammatory response. Moreover, arsenic-exposed cells displayed persistent DNA damage, as indicated by increased γ-H2AX foci, accompanied by nuclear structural alterations and elevated expression of cancer stem cell markers, including OCT2, CD133, and ALDH1. These findings suggest that arsenic-induced inflammation and genomic instability converge to promote a tumor-supportive microenvironment, highlighting the potential role of chronic arsenic exposure in ovarian carcinogenesis, particularly in the context of inflammation-driven carcinogenesis.
Collapse
Affiliation(s)
- Cristina M Andrade-Feraud
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Arlet M Acanda de la Rocha
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Noah E Berlow
- First Ascent Biomedical, Inc., United States of America
| | - Santiago Duque
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Alexander Velazco
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Diego Castillo
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Baylee Holcomb
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Ebony R Coats
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Yasmin R Ghurani
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Catherine M Lucey
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, United States of America
| | - Brandon Pearson
- Environmental and Molecular Toxicology, Oregon State University, OR, United States of America
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America
| | - Diana J Azzam
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States of America.
| |
Collapse
|
12
|
Rosenbaum SR, Hughes CJ, Fields KM, Purdy SC, Gustafson AL, Wolin A, Hampton D, Shrivastava NM, Turner N, Danis E, Ebmeier C, Spoelstra N, Richer J, Jedlicka P, Costello JC, Zhao R, Ford HL. EYA3 regulation of NF-κB and CCL2 suppresses cytotoxic NK cells in the premetastatic niche to promote TNBC metastasis. SCIENCE ADVANCES 2025; 11:eadt0504. [PMID: 40333987 PMCID: PMC12057687 DOI: 10.1126/sciadv.adt0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
Triple-negative breast cancer cells must evade immune surveillance to metastasize to distant sites, yet this process is not well understood. The Eyes absent (EYA) family of proteins, which are crucial for embryonic development, become dysregulated in cancer, where they have been shown to mediate proliferation, migration, and invasion. Our study reveals an unusual mechanism by which EYA3 reduces the presence of cytotoxic natural killer (NK) cells in the premetastatic niche (PMN) to enhance metastasis, independent of its effects on the primary tumor. We find that EYA3 up-regulates nuclear factor κB signaling to enhance CCL2 expression, which, in contrast to previous findings, suppresses cytotoxic NK cell activation in vitro and their infiltration into the PMN in vivo. These findings uncover an unexpected role for CCL2 in inhibiting NK cell responses at the PMN and suggest that targeting EYA3 could be an effective strategy to reactivate antitumor immune responses to inhibit metastasis.
Collapse
Affiliation(s)
- Sheera R. Rosenbaum
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Connor J. Hughes
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Kaiah M. Fields
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Stephen Connor Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Annika L. Gustafson
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Arthur Wolin
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Drake Hampton
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Natasha M. Shrivastava
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Nicholas Turner
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Etienne Danis
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Christopher Ebmeier
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Nicole Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Jennifer Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Paul Jedlicka
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Rui Zhao
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| |
Collapse
|
13
|
Hasan R, Hasan MM, Shathi JH, Tamam E, Ahmed AE, Haque A, Rahmann Z, Islam MT, Reza MA, Biswas MS, Hoque KMF. Toxic effects of malachite green on plant and animal models: A study on root growth inhibition, hematological changes, histopathology, and molecular analysis. Toxicol Lett 2025; 409:61-73. [PMID: 40348348 DOI: 10.1016/j.toxlet.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/03/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Malachite green (MG), a suggestive chemical for tumor development and carcinogenicity, is widely used as an illicit food coloring agent, posing risks to consumers and handlers. This study aimed to assess the toxic effects of MG in both plant and animal models. Different doses of MG (375, 750, and 1500 mg/L) were applied for 24 h to evaluate root growth inhibition, mitotic index (MI), and chromosomal aberrations in Allium cepa L. roots for genotoxicity analysis. In animal studies, forty Swiss albino mice were divided into four groups: control and three treatment groups, which were orally administered MG at 375 (low), 750 (medium), and 1500 (high) mg/kg body weight for 13 weeks. Hematological, biochemical, histopathological, and molecular analyses were performed on liver, kidney, and intestinal tissues post-treatment. MG significantly reduced root length and MI in A. cepa roots dose-dependently causing chromosomal abnormalities. MG treatment significantly lowered the body weights of mice and increased platelet, monocyte, and white blood cell counts, while reducing hemoglobin, hematocrit, and red blood cell counts. Serum analysis showed elevated ALT, ALP, AST, bilirubin, creatinine, and urea, indicating hepatotoxicity and nephrotoxicity. Histopathological examination revealed vacuolation, congestion, and inflammatory infiltration in the liver, glomerular shrinkage, tubular degeneration, and interstitial edema in the kidney, and epithelial sloughing, submucosal necrosis, and inflammatory infiltration in the colon. RT-qPCR analysis demonstrated increased Bcl-2, Beclin-1, and NF-κB mRNA expression with decreased Bax mRNA. These findings suggest MG is a potent genotoxic and carcinogenic agent even at lower doses threatening human health.
Collapse
Affiliation(s)
- Rubait Hasan
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh; Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Mahmudul Hasan
- Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Jamiatul Husna Shathi
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh; Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Esraa Tamam
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Egypt
| | - Amaal E Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Aswan University, Egypt
| | - Ariful Haque
- Molecular Pathology Laboratory, Institute of Biological Sciences, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Zahidur Rahmann
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh; Riceland Healthcare, 538 Broadway Ave, Winnie, TX 77665, USA
| | - Md Tariqul Islam
- Department of Biochemistry & Biotechnology, School of Biomedical Science, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Md Abu Reza
- Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Mohammad Shahangir Biswas
- Department of Biochemistry and Biotechnology, University of Science & Technology Chittagong (USTC), Foy's Lake, Zakir Hossain Road, Chittagong 4202, Bangladesh; Department of Public Health, Daffodil International University, Dhaka 1216, Bangladesh.
| | - Kazi Md Faisal Hoque
- Molecular Biology and Protein Science Laboratory, Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| |
Collapse
|
14
|
Yang W, Wang F, Liu J, Wang X, Zhang H, Gao D, Wang A, Jin Y, Chen H. β-Hydroxybutyrate aggravates LPS-induced inflammatory response in bovine endometrial epithelial cells by activating the oxidative stress/NF-κB signaling pathway. Int Immunopharmacol 2025; 154:114609. [PMID: 40188524 DOI: 10.1016/j.intimp.2025.114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/08/2025]
Abstract
Ketosis, a metabolic disorder characterized by elevated levels of ketone bodies in the blood or urine, is known to impair the health and productivity of dairy cows, leading to substantial economic losses in the dairy industry. When ketosis occurs in dairy cows, the levels of β-hydroxybutyrate (BHBA), an abundant form of ketone bodies, in the blood increase significantly. Elevated BHBA levels have been shown to negatively impact reproductive performance and increase the incidence of periparturient diseases in dairy cows, including mastitis and endometritis. However, the role of BHBA in the development of endometritis in dairy cows and its underlying mechanisms remain largely unclear. The present study was designed to investigate the specific role of BHBA in the development of endometritis using an inflammatory response model of the bovine endometrial epithelial cell line (BENDs). Escherichia coli lipopolysaccharide (LPS) treatment (1 μg/mL) significantly increased the expression levels of interleukin (IL)-6 and IL-1β, as well as the phosphorylation of p65 and IκB in BENDs. In addition, co-treatment with BHBA (2.4 mM) and LPS (1 μg/mL) significantly increased the expression levels of proinflammatory cytokines (IL-6, IL-1β, and IL-8), as well as the phosphorylation of p65 and IκB, compared to the LPS-only treatment group. Immunofluorescence staining showed that the addition of LPS altered the nuclear localization of p65, and co-treatment with BHBA and LPS further promoted the translocation of p65 to the nucleus. Additionally, the addition of BHBA significantly increased the levels of oxidation indicators (MDA), whereas the levels of antioxidative indicators, including heme oxygenase-1 (HO-1) and catalase (CAT), were markedly decreased in BENDs. As a representative antioxidant, N-acetylcysteine (NAC) treatment significantly reduced the phosphorylation of p65 and IκB in the BHBA and LPS co-treatment group. SC75741, an NF-κB signaling pathway inhibitor, significantly decreased the expression levels of proinflammatory cytokines (IL-6, IL-1β, IL-8, and CCL5) in the BHBA and LPS co-treatment group. In summary, the current study demonstrates that BHBA aggravates LPS-induced inflammatory response in BENDs through the activation of oxidative stress/NF-κB signaling pathway, unravelling the mechanism by which BHBA exacerbates the inflammatory response in the BENDs of dairy cattle. This study elucidates the role of ketosis and its key metabolite BHBA in the pathogenesis of endometritis in dairy cows, providing valuable insights for understanding this pathological process.
Collapse
Affiliation(s)
- Wanghao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Fengbo Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jijun Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xuerong Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haisen Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Dengke Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China; Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
15
|
Stutts J, Clatterbuck K, Duckworth C, Pemberton T, Elkins A, Patra P, Stoecker W, Geria N, Nosoudi N. Synergistic impact of antioxidant combinations on collagen and elastin synthesis in human dermal fibroblasts. Biomed Mater Eng 2025:9592989251341159. [PMID: 40340419 DOI: 10.1177/09592989251341159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
BACKGROUND The restoration of collagen and elastin in human dermal fibroblasts plays a crucial role in anti-aging and skin rejuvenation therapies. Numerous studies have examined the effects of various antioxidants on skin health, but there is limited research comparing their combined effects on collagen and elastin synthesis in human dermal fibroblasts. Objective: The objective of this study was to evaluate the individual and combined effects of N-acetylcysteine (NAC), Coenzyme Q10 (CoQ10), Niacinamide (NIAC), Gamma Cyclodextrin (GAMMA), Retinol (RET), Epigallocatechin Gallate (EGCG), and Ellagic Acid (ELA) on collagen type I and elastin synthesis in human dermal fibroblasts (HDFs). Methods: Human dermal fibroblasts were treated with individual and combined antioxidants. The expression of collagen type I and elastin was measured using mRNA analysis, immunofluorescence staining, and matrix protein assays. The study focused on the effects of EGCG in combination with other antioxidants like RET, CoQ10, and NAC to identify synergistic effects. Results: The combination of EGCG + RET and EGCG + CoQ10 showed the most significant increase in both elastin and collagen type I synthesis, surpassing the effects of individual antioxidants. EGCG demonstrated the highest fold change in elastin mRNA expression, while the combination treatments notably enhanced the extracellular matrix restoration in HDFs. Conclusion: The combination of EGCG with CoQ10, Retinol, or NAC presents a promising strategy for enhancing skin elasticity and firmness by promoting both elastin and collagen synthesis. These findings suggest that antioxidant combinations can be developed for effective anti-aging skincare formulations.
Collapse
Affiliation(s)
- Jada Stutts
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| | - Kayla Clatterbuck
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| | - Chloe Duckworth
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| | - Tyera Pemberton
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| | - Aillea Elkins
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| | - Prabir Patra
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| | - William Stoecker
- Department of Dermatology, University of Missouri, Columbia, MO, USA
| | - Navin Geria
- S&A Technologies, Rolla, MO, USA
- AyurDerm Technologies LLC, Warren, NJ, USA
| | - Nasim Nosoudi
- Department of Biomedical Engineering, College of Engineering and Computer Sciences, Marshall University, Huntington, WV, USA
| |
Collapse
|
16
|
Liu C, Peng C, Jia X, Yan C, Liu D, Zhang X, Song H, Han Y. Determining the biomarkers and pathogenesis of myocardial infarction combined with ankylosing spondylitis via a systems biology approach. Front Med 2025:10.1007/s11684-025-1132-8. [PMID: 40317453 DOI: 10.1007/s11684-025-1132-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 05/07/2025]
Abstract
Ankylosing spondylitis (AS) is linked to an increased prevalence of myocardial infarction (MI). However, research dedicated to elucidating the pathogenesis of AS-MI is lacking. In this study, we explored the biomarkers for enhancing the diagnostic and therapeutic efficiency of AS-MI. Datasets were obtained from the Gene Expression Omnibus database. We employed weighted gene co-expression network analysis and machine learning models to screen hub genes. A receiver operating characteristic curve and a nomogram were designed to assess diagnostic accuracy. Gene set enrichment analysis was conducted to reveal the potential function of hub genes. Immune infiltration analysis indicated the correlation between hub genes and the immune landscape. Subsequently, we performed single-cell analysis to identify the expression and subcellular localization of hub genes. We further constructed a transcription factor (TF)-microRNA (miRNA) regulatory network. Finally, drug prediction and molecular docking were performed. S100A12 and MCEMP1 were identified as hub genes, which were correlated with immune-related biological processes. They exhibited high diagnostic value and were predominantly expressed in myeloid cells. Furthermore, 24 TFs and 9 miRNA were associated with these hub genes. Enzastaurin, meglitinide, and nifedipine were predicted as potential therapeutic agents. Our study indicates that S100A12 and MCEMP1 exhibit significant potential as biomarkers and therapeutic targets for AS-MI, offering novel insights into the underlying etiology of this condition.
Collapse
Affiliation(s)
- Chunying Liu
- Beifang Hospital of China Medical University, Shenyang, 110016, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China
| | - Chengfei Peng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China
| | - Xiaodong Jia
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China
| | - Dan Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China
| | - Xiaolin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China
| | - Haixu Song
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China.
| | - Yaling Han
- Beifang Hospital of China Medical University, Shenyang, 110016, China.
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Cardiovascular Research Institute and Department of Cardiology, Shenyang, 110016, China.
| |
Collapse
|
17
|
Holm H, Magnusson M, Jujić A, Lagrange J, Bozec E, Lamiral Z, Bresso E, Huttin O, Baudry G, Monzo L, Rossignol P, Zannad F, Girerd N. Association of ventricular-arterial coupling with biomarkers involved in heart failure pathophysiology - the STANISLAS cohort. Eur J Heart Fail 2025; 27:860-871. [PMID: 39189882 DOI: 10.1002/ejhf.3411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
AIMS Impaired left ventricular-arterial coupling (VAC) has been shown to correlate with worse prognosis in cardiac diseases and heart failure (HF). The extent of the relationship between VAC and circulating biomarkers associated with HF has been scarcely documented. We aimed to explore associations of VAC with proteins involved in HF pathophysiology within a large population-based cohort of middle-aged individuals. METHODS AND RESULTS In the forth visit of the STANISLAS family cohort, involving 1309 participants (mean age 48 ± 14 years; 48% male) from parent and children generations, we analysed the association of 32 HF-related proteins with non-invasively assessed VAC using pulse wave velocity (PWV)/global longitudinal strain (GLS) and arterial elastance (Ea)/ventricular end-systolic elastance (Ees). Among the 32 tested proteins, fatty acid-binding protein adipocyte 4, interleukin-6, growth differentiation factor 15, matrix metalloproteinase (MMP)-1, and MMP-9 and adrenomedullin were positively associated with PWV/GLS whereas transforming growth factor beta receptor type 3, MMP-2 and N-terminal pro-B-type natriuretic peptide (NT-proBNP) were negatively associated. In multivariable models, only MMP-2 and NT-proBNP were significantly and inversely associated with PWV/GLS in the whole population and in the parent generation. Higher levels of NT-proBNP were also negatively associated with Ea/Ees in the whole cohort but this association did not persist in the parent subgroup. CONCLUSION Elevated MMP-2 and NT-proBNP levels correlate with better VAC (lower PWV/GLS), possibly indicating a compensatory cardiovascular response to regulate left ventricular pressure amidst cardiac remodelling and overload.
Collapse
Affiliation(s)
- Hannes Holm
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Martin Magnusson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Amra Jujić
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Jérémy Lagrange
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Erwan Bozec
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Zohra Lamiral
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Emmanuel Bresso
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Olivier Huttin
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Guillaume Baudry
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Luca Monzo
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| |
Collapse
|
18
|
Osburn SC, Smith ME, Wahl D, LaRocca TJ. Novel effects of reverse transcriptase inhibitor supplementation in skeletal muscle of old mice. Physiol Genomics 2025; 57:308-320. [PMID: 40062980 DOI: 10.1152/physiolgenomics.00115.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/08/2024] [Accepted: 02/15/2025] [Indexed: 04/01/2025] Open
Abstract
Aging is the primary risk factor for the development of many chronic diseases, including dementias, cardiovascular disease, and diabetes. There is significant interest in identifying novel "geroprotective" agents, including by repurposing existing drugs, but such treatments may affect organ systems differently. One current example is the nucleoside reverse transcriptase inhibitor 3TC, which has been increasingly studied as a potential gerotherapeutic. Recent data suggest that 3TC may reduce inflammation and improve cognitive function in older mice; however, the effects of 3TC on other tissues in aged animals are less well characterized. Here, we use transcriptomics (RNA-seq) and targeted metabolomics to investigate the influence of 3TC supplementation on skeletal muscle in older mice. We show that 3TC 1) does not overtly affect muscle mass or functional/health markers, 2) largely reverses age-related changes in gene expression and metabolite signatures, and 3) is potentially beneficial for mitochondrial function in old animals via increases in antioxidant enzymes and decreases in mitochondrial reactive oxygen species. Collectively, our results suggest that, in addition to its protective effects in other tissues, 3TC supplementation does not have adverse effects in aged muscle and may even protect muscle/mitochondrial health in this context.NEW & NOTEWORTHY Recent studies suggest that the nucleoside reverse transcriptase inhibitor 3TC may improve brain health and cognitive function in old mice, but its effects on other aging tissues have not been comprehensively studied. This is the first study to use a multiomics approach to investigate the effects of 3TC treatment on skeletal muscle of old mice. The results suggest that 3TC reverses age-related transcriptomic and metabolite signatures and is potentially beneficial for mitochondrial function in aged muscle.
Collapse
Affiliation(s)
- Shelby C Osburn
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Meghan E Smith
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Devin Wahl
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| | - Thomas J LaRocca
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States
| |
Collapse
|
19
|
Liu X, Lin Y, Zhuang Q, Deng H, Liu A, Sun J. BTK inhibitors resistance in B cell malignancies: Mechanisms and potential therapeutic strategies. Blood Rev 2025; 71:101273. [PMID: 40000280 DOI: 10.1016/j.blre.2025.101273] [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: 11/17/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Bruton tyrosine kinase inhibitors (BTKi) have shown prominent clinical efficacy in patients with B cell malignancies, such as chronic lymphocytic leukemia, mantle cell lymphoma, diffuse large B cell lymphoma, and Waldenström's macroglobulinemia. Nevertheless, numerous factors contribute to BTKi resistance, encompassing genetic mutations, chromosomal aberrations, dysregulation of protein expression, tumor microenvironment, and metabolic reprogramming. Accordingly, potential therapeutic strategies have been explored to surmount BTKi resistance, including noncovalent BTKi, BTK proteolysis-targeting chimeras, and combination therapies. Herein, we summarize the mechanisms responsible for BTKi resistance as well as the current preclinical and clinical strategies to address BTKi resistance in B cell malignancies treatment.
Collapse
Affiliation(s)
- Xin Liu
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yufan Lin
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiqi Zhuang
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoren Deng
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China
| | - Aichun Liu
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Jie Sun
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, China.
| |
Collapse
|
20
|
Liu X, Wang H, Yuan M, Zhang T, Wang Q, Chen N, Zhou X, He M, Ji Z, Shen H. m 6A-modified RIOK3 activated the NF-κB-signaling pathway by CDC42, promoting the replication and proliferation of enterovirus. Int J Biol Macromol 2025; 305:140988. [PMID: 39961559 DOI: 10.1016/j.ijbiomac.2025.140988] [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: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Enterovirus infections are implicated in the pathogenesis of inflammatory diseases, such as viral myocarditis, meningitis, and pancreatitis. These infections activate innate and inflammatory immune responses upon viral entry into host cells. However, the precise mechanisms through which enteroviruses induce inflammation to facilitate viral replication remain unclear. N(6)-methyladenosine (m6A), one of the most abundant internal modifications on eukaryotic mRNAs, is regulated by METTL3, a key "writer" enzyme in the m6A methyltransferase complex. This study identifies RIO kinase 3 (RIOK3), a serine-threonine protein kinase, involved in innate immunity, inflammation, and cell cycle regulation, as a critical factor in Coxsackievirus B3 (CVB3) infection. CVB3 infection significantly increases RIOK3 expression both in vivo and in vitro, accompanied by elevated m6A modifications on RIOK3 mRNA. METTL3-mediated m6A modification enhances RIOK3 transcription, which in turn downregulates CDC42, a small GTPase of the Rho subfamily, that regulates key cellular processes, including antiviral signaling. This suppression of CDC42 promotes CVB3 replication. Additionally, RIOK3 and CDC42 modulate the NF-κB signaling pathway, a pivotal regulator of inflammatory and immune responses during infection. These findings reveal that m6A-modified RIOK3 promotes enterovirus replication by activating the NF-κB signaling pathway via CDC42 suppression, providing novel insights into the molecular mechanisms of enterovirus pathogenesis.
Collapse
Affiliation(s)
- Xiaolan Liu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China; Department of Clinical Laboratory, Norinco General Hospital, Xi'an, Shaanxi 710065, PR China
| | - Hua Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Mengran Yuan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Tianyi Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Qimeng Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Nuo Chen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Xiaoxiang Zhou
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Min He
- Nanjing Center for Disease Control and Prevention, Nanjing 210003, PR China
| | - Zengjun Ji
- Department of Laboratory Medicine, Taizhou Second People's Hospital, Taizhou 225599, PR China.
| | - Hongxing Shen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
21
|
Yu Y, Li J, Wang X, Li X, Lyu C, Yang L, Bai Y. Actin Gamma Smooth Muscle 2 Promotes Epithelial Ovarian Cancer Cell Proliferation via the AKT1/NF-κB Signaling Pathway. Cell Biochem Funct 2025; 43:e70077. [PMID: 40289545 DOI: 10.1002/cbf.70077] [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: 10/07/2024] [Revised: 02/15/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
Epithelial ovarian cancer (EOC) is associated with high mortality rates worldwide and is characterized as the most lethal gynecological cancer. The study aimed to investigate the functional role and underlying molecular mechanism of actin gamma smooth muscle 2 (ACTG2) in the progression of EOC. Data mining from The Cancer Genome Atlas (TCGA) databases showed the expression of ACTG2 was significantly upregulated in EOC and negatively associated with longer overall survival and better prognosis of patients. By using of gain-of-function and loss-of-function experiments in vitro and in vivo, we found that ACTG2 promoted EOC cell proliferation and suppressed cell apoptosis. Mechanistic study revealed that ACTG2 regulates EOC cell proliferation by activating the AKT serine/threonine kinase 1 (AKT1)/nuclear factor-κB (NF-κB) signaling pathway. Importantly, p65 plays a crucial role in this newly identified regulatory mechanism. Our findings demonstrate that ACTG2 may play an oncogenic role in EOC, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yinjue Yu
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jiangxia Li
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xiaohang Wang
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxiao Li
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Cuiting Lyu
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Lina Yang
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yongrui Bai
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Castel T, Pichavant‐Rafini K, Théron M, Gandubert C, Léon K. Effects of selenium, selenium-enriched spirulina and phycocyanin on myocarditis stress parameters on LPS-induced injury in H9c2 ventricular cardiomyoblasts. Physiol Rep 2025; 13:e70376. [PMID: 40387469 PMCID: PMC12087287 DOI: 10.14814/phy2.70376] [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: 02/18/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Myocarditis is strongly represented in septic patients and is associated with a higher mortality rate. Spirulina platensis (Spi), a blue-green algae, has anti-inflammatory properties and can be enriched with selenium, an antioxidant essential oligoelement. In addition, phycocyanin (PC), a biliprotein extract from spirulina, displays interesting anti-inflammatory and antiapoptotic effects. In this study, the objective was to determine the cardioprotective effects of Sodium selenite (Se), Spi, Spi + Se (SeSP) and PC on LPS-induced inflammation, apoptosis, and oxidative stress parameters. H9c2 cells were co-treated with or without LPS (5 μg/mL) and Se (0.5 μM), Spi (2.5 μg/mL), SeSP (0.5 μM Se + 2.5 μg/mL Spi) and PC (0.1 μg/mL) for 24 h. Inflammation was investigated by measurement of NFκB activation, IL-6, and caspase 1 expression, while apoptosis was measured by Bax, Bcl-2, and caspase-3 expression. Furthermore, GPx and SOD activities were analyzed, as well as isoprostanes and nrf-2 expression. Activation of MAPK Junk and p38 was also determined. Our results demonstrated that Se could only reduce p65 S536 phosphorylation. SeSP could limit Bax expression, while an increase in IL-6 was detected without LPS. Moreover, PC could reduce IL-6 and Caspase-1 expression and could have promising properties to decrease LPS-induced myocarditis outcomes.
Collapse
|
23
|
Li Y, Hong Y, Shen H, Zhou J, Cesar D, Eleutério J, Matsuura M, Liu Y, Luo C, Li Q. FXR activation suppresses NF-κB signaling, proliferation and migration in cervical cancer cells. Transl Cancer Res 2025; 14:2440-2456. [PMID: 40386262 PMCID: PMC12079256 DOI: 10.21037/tcr-2025-522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
Background The Farnesoid X receptor (FXR) is a nuclear receptor known for its role in inflammation regulation and tumor suppression in various cancers. However, its functional significance and underlying mechanisms in cervical cancer (CC) remain unclear. The persistent activation of the nuclear factor kappa B (NF-κB) signaling pathway due to inflammation is a key driver of cancer progression. This study investigates the effects of FXR activation in CC and its interaction with the NF-κB pathway. Methods CC cells were treated with GW4064, an FXR agonist (3 µM), and xenograft tumor models were assigned to receive 30 mg/kg GW4064. NF-κB-mediated transcriptional activity was assessed using a dual-luciferase reporter assay. Gene expression in CC cells and mouse tissues was analyzed via quantitative real-time polymerase chain reaction (qRT-PCR), while key proteins in the NF-κB and STAT3 signaling pathways were examined using Western blotting. Cell proliferation, migration, and invasion were evaluated through methylthiazolyldiphenyl-tetrazolium bromide (MTT), wound healing, and real-time cellular analysis (RTCA), respectively. Apoptosis was measured using a fluorescein isothiocyanate (FITC) Annexin V Apoptosis Detection Kit I. Results FXR deletion in 6- to 8-week-old C57B/6 female mice led to abnormal upregulation of inflammatory genes in the cervix and aberrant NF-κB activation. Treatment with GW4064 suppressed NF-κB-regulated gene expression in Hela and Siha CC cells and inhibited NF-κB activity at the transcriptional level. Mechanistically, FXR activation suppressed tumor necrosis factor alpha (TNFα)-induced phosphorylation of NF-κB inhibitor alpha (IκBα) by directly binding to the promoter of inhibitor of nuclear factor kappa B kinase regulatory subunit gamma (IKBKG), thereby inhibiting its transcription. Additionally, FXR activation reduced CC cell proliferation and migration. In vivo, xenograft experiments in Hela cell-bearing Bagg's albino (BALB/c) nude female mice confirmed that FXR activation significantly suppressed tumor growth. Conclusions These findings highlight FXR activation as a potential therapeutic strategy for CC by targeting the NF-κB pathway as shown in both in vitro and in vivo.
Collapse
Affiliation(s)
- Yuanqiang Li
- Key Laboratory of Integrated Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | | | - Huize Shen
- Key Laboratory of Integrated Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jingnan Zhou
- Procurement Department, Zhejiang Cancer Hospital, Hangzhou, China
| | - Daniel Cesar
- Gynecology Oncology-National Cancer Institute, Rio de Janeiro, Brazil
| | - José Eleutério
- Department of Women, Children and Adolescent Health, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Motoki Matsuura
- Department of Obstetrics and Gynecology, Sapporo Medical University, Sapporo, Japan
| | | | - Cong Luo
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Qinglin Li
- Key Laboratory of Integrated Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Kumar A, Bharadwaj T, Muthuraj L, Kumar J, Kumar P, Lalitha R, Sigamani G, Ahmad S, Bhanu P, Pathak RK, Uttarkar A, Niranjan V, Mishra V. Molecular dynamics simulation and docking studies reveals inhibition of NF-kB signaling as a promising therapeutic drug target for reduction in cytokines storms. Sci Rep 2025; 15:15225. [PMID: 40307269 PMCID: PMC12043994 DOI: 10.1038/s41598-024-78411-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/30/2024] [Indexed: 05/02/2025] Open
Abstract
Nuclear factor-kappa B (NF-kB) plays a crucial role in numerous cellular processes, such as inflammation, immunological responses to infection, cell division, apoptosis, and the development of embryos and neurons. Cytokines, plays an important role in positive feedback loop and leads to inflammatory cell death through the release of pathogenic cytokine known to be cytokine storm which causes diseases like Acute Respiratory Disorder (ARD), multi-organ disorder, Hyperinflammation syndrome and may cause death. This cytochrome storm was identified in the people severely affected by covid-19. NF-kB presents a promising therapeutic opportunity to mitigate covid-19-induced cytokine storm and reduce the risk of severe morbidity and mortality resulting from the diseases. This paper therefore explores the modulation of the NF-kB pathway by inhibiting the binding of the transcription factor as a potential strategy to mitigate the morbidity and mortality caused by cytokine storms. To identify small molecule inhibitors of NF-kB signaling, we screened approximately 101 molecules in two identified pockets of NF-kB (p50/p65)-DNA complex. Each molecule was virtually screened in two pockets (A1 and A2). The focus library was developed starting from chemical structures obtained from the literature (Angelicin and Psolaren) which shows the inhibition of NF-kB signaling, as well as using artificial intelligence (WADDAICA) and rationally designed molecules. Among the 3 highest-scored ligands (NFAI64, NF30 and NF49) selected from the docking studies and further molecular dynamic investigations. The identified compound NF30 showed significantly higher binding affinity (ΔGbinding) in A2 pocket (60.92 ± 1.83 kJ/mol) as compared to the rest of the molecules, making it a promising molecule for the inhibition of NF-kB. The discovered novel compounds by computational studies could be of relevance to identify more potent inhibitors of NF-kB dependent biological functions beneficial to control the cytokine storm occurring in the patients affected with Covid-19.
Collapse
Affiliation(s)
- Abhishek Kumar
- Department of Computational Biology and AI, Kcat Enzymatic Private Limited, #16, Ramakrishnappa Road, Cox Town, Bangalore, 560005, India
| | - Tharun Bharadwaj
- Department of Computational Biology and AI, Kcat Enzymatic Private Limited, #16, Ramakrishnappa Road, Cox Town, Bangalore, 560005, India
| | - Likith Muthuraj
- Department of Computational Biology and AI, Kcat Enzymatic Private Limited, #16, Ramakrishnappa Road, Cox Town, Bangalore, 560005, India
| | - Jitendra Kumar
- Biotechnology Industry Research Assistance Council (BIRAC), NSIC Business Park, NSIC Bhawan, Okhla Industrial Estate, New Delhi, 110020, India.
| | - Pravin Kumar
- Department of Computational Biology and AI, Kcat Enzymatic Private Limited, #16, Ramakrishnappa Road, Cox Town, Bangalore, 560005, India.
| | - Roopa Lalitha
- Department of Computational Biology and AI, Kcat Enzymatic Private Limited, #16, Ramakrishnappa Road, Cox Town, Bangalore, 560005, India
| | - Gladstone Sigamani
- Department of Computational Biology and AI, Kcat Enzymatic Private Limited, #16, Ramakrishnappa Road, Cox Town, Bangalore, 560005, India
| | - Shaban Ahmad
- International Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Piyush Bhanu
- Xome Life Sciences, Bangalore Bioinnovation Center, Helix Biotech Park, Bangalore, 560100, India
| | - Ravi Kant Pathak
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Delhi GT Rd, Phagwara, Punjab, 144001, India
| | - Akshay Uttarkar
- Department of Biotechnology, RV College of Engineering, RV Vidyanikethan Post, Mysuru Road, Banglore, 560059, India
| | - Vidya Niranjan
- Department of Biotechnology, RV College of Engineering, RV Vidyanikethan Post, Mysuru Road, Banglore, 560059, India
| | - Vachaspati Mishra
- Department of Botany, Dyal Singh College, University of Delhi, Delhi, 110003, India
| |
Collapse
|
25
|
Bai YS, Wang DL, Lee MC, Wang CC, Fang WH, Chuang SW, Chen YH, Su H, Chen CJ, Su SL. Dissect Gender-Dependent Susceptibility SNPs in Progressive Osteoarthritis Using Regulator Motif Candidate of Genetic Association Strategy (RMCGA). Int J Mol Sci 2025; 26:4117. [PMID: 40362356 PMCID: PMC12071535 DOI: 10.3390/ijms26094117] [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: 03/25/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
The role of gender in osteoarthritis (OA) has been reported. However, knowledge on whether gender-specific regulatory SNPs are determining factors in OA is limited. We aimed to identify susceptible gender-specific SNPs of transcription factor binding sites in OA. We used a modified NF-κB binding motif from an RNA sequencing data-inferred OA-associated upstream regulator to define genome-wide potential NF-κB binding sites, which were aligned to the Taiwan BioBank SNP database to identify susceptible SNPs. A case-control study was conducted to verify SNPs with OA determined by a logistic model. The functional assessment was validated using the Genotype-Tissue Expression Portal database. We collected 533 OA patients and 614 healthy controls. Two of nine novel OA-associated SNPs were identified to be significant. For males, the variant of rs73164856 in the aldose reductase gene enhancer was identified to be a protective factor of severe OA patients [odds ratio (OR): 0.17, 95% confidence interval (CI): 0.04-0.73]. For females, the variant of the rs545654 in the neuronal NOS (nNOS) gene was identified to be a detrimental factor of severe OA patients (OR: 2.07, 95% CI: 1.15-3.73). The gene expression analysis demonstrated a lower expression of the AKR1B15 gene (p = 0.00019) upon the rs73164856 T allele; meanwhile, it showed a higher expression of the nNOS gene (p = 1.2 × 10-17) upon the rs545654 T allele. This study identifies susceptible gender-specific SNPs of NF-κB binding sites in severe OA and validates the RMCGA, which sheds light on genetic determinants by gender in advanced OA.
Collapse
Affiliation(s)
- Yin-Shiuan Bai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (Y.-S.B.); (D.-L.W.)
| | - Ding-Lian Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (Y.-S.B.); (D.-L.W.)
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Meng-Chang Lee
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Chih-Chien Wang
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan;
- School of Medicine, National Defense Medical Center, Taipei 114201, Taiwan;
| | - Wen-Hui Fang
- School of Medicine, National Defense Medical Center, Taipei 114201, Taiwan;
- Department of Family and Community Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Su-Wen Chuang
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Yu-Hsuan Chen
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Hao Su
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan
| | - Cheng-Jung Chen
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan;
- Taichung Veterans General Hospital Chiayi Branch, Chiayi City 60090, Taiwan
| | - Sui-Lung Su
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (Y.-S.B.); (D.-L.W.)
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| |
Collapse
|
26
|
Hameed SA, Kolch W, Brennan DJ, Zhernovkov V. Direct cell interactions potentially regulate transcriptional programmes that control the responses of high grade serous ovarian cancer patients to therapy. Sci Rep 2025; 15:14484. [PMID: 40280979 PMCID: PMC12032223 DOI: 10.1038/s41598-025-98463-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
The tumour microenvironment is composed of a complex cellular network involving cancer, stromal and immune cells in dynamic interactions. A large proportion of this network relies on direct physical interactions between cells, which may impact patient responses to clinical therapy. Doublets in scRNA-seq are usually excluded from analysis. However, they may represent directly interacting cells. To decipher the physical interaction landscape in relation to clinical prognosis, we inferred a physical cell-cell interaction (PCI) network from 'biological' doublets in a scRNA-seq dataset of approximately 18,000 cells, obtained from 7 treatment-naive ovarian cancer patients. Focusing on cancer-stromal PCIs, we uncovered molecular interaction networks and transcriptional landscapes that stratified patients in respect to their clinical responses to standard therapy. Good responders featured PCIs involving immune cells interacting with other cell types including cancer cells. Poor responders lacked immune cell interactions, but showed a high enrichment of cancer-stromal PCIs. To explore the molecular differences between cancer-stromal PCIs between responders and non-responders, we identified correlating gene signatures. We constructed ligand-receptor interaction networks and identified associated downstream pathways. The reconstruction of gene regulatory networks and trajectory analysis revealed distinct transcription factor (TF) clusters and gene modules that separated doublet cells by clinical outcomes. Our results indicate (i) that transcriptional changes resulting from PCIs predict the response of ovarian cancer patients to standard therapy, (ii) that immune reactivity of the host against the tumour enhances the efficacy of therapy, and (iii) that cancer-stromal cell interaction can have a dual effect either supporting or inhibiting therapy responses.
Collapse
Affiliation(s)
- Sodiq A Hameed
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland.
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| | - Donal J Brennan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
- UCD Gynaecological Oncology Group Catherine McAuley Research Centre, Mater Misericordiae University Hospital, Eccles Street, Dublin, D07 R2WY, Ireland
| | - Vadim Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| |
Collapse
|
27
|
Lee M, Park J, Kim D, Park SH, Jung J, Jun W, Kim J, Baek KS, Kim OK, Lee J. Effect of water extract of bay laurel ( Laurus nobilis L.) on non-alcoholic fatty liver disease (NAFLD). Food Nutr Res 2025; 69:10668. [PMID: 40313587 PMCID: PMC12045078 DOI: 10.29219/fnr.v69.10668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/10/2024] [Accepted: 08/30/2024] [Indexed: 05/03/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) involves lipid accumulation in liver without consumption of alcohol and affects many people worldwide. NAFLD is associated with metabolic syndrome disease such as obesity, insulin resistance, hyperlipidemia, and diabetes. However, there are no pharmacologic therapies for NAFLD. Recently, there are increasing reports that several natural plants can inhibit lipid accumulation in hepatocytes. Bay laurel (Laurus nobilis L.) leaves have been used in traditional medicine for rheumatism, stomach ache, emetic, skin rashes, and earaches. Our objective was to investigate the effect of bay laurel leaves water extract (BLW) on free fatty acid (FFA) treated hepatocyte and high fructose, high fat (HFHF) diet in a mouse model of NAFLD. In vitro, lipid accumulation increased only in the FFA treated group, while BLW reduced lipid accumulation to a level comparable to that only in the FFA treated group. Cellular antioxidants were increased in the BLW compared to the only FFA-treated group, but cellular MDA levels were decreased in the BLW compared to the only FFA treated group. Cellular lipid accumulation, inflammation, and apoptosis were reduced in the BLW compared to the only FFA treated group. In vivo, serum ALT, AST, and GGT levels in the BLW supplementation group were significantly decreased compared with the HFHF group. Hepatic TC, TG, and MDA levels were significantly decreased in the HFHF+100 and HFHF+200 groups compared to the HFHF group. The hepatic antioxidant activities in the BLW supplementation groups were significantly increased compared to the HFHF group. The expression of proteins related to hepatic inflammation and apoptosis was reduced in the BLW supplementation groups compared to the HFHF group. These results suggest that BLW could be potentially useful in the treatment of NAFLD due to its inhibitory effects on hepatic lipogenesis, hepatic inflammation, and hepatic apoptosis.
Collapse
Affiliation(s)
- Minhee Lee
- Department of Food Innovation and Health, Kyung Hee University, Yongin 17104, Korea
| | - Jeongjin Park
- Division of Food and Nutrition and Human Ecology Research Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dakyung Kim
- Department of Medical Nutrition, Kyung Hee University, Yongin 17104, Korea
| | - Seong-Hoo Park
- Department of Medical Nutrition, Kyung Hee University, Yongin 17104, Korea
| | - Jaeeun Jung
- Department of Medical Nutrition, Kyung Hee University, Yongin 17104, Korea
| | - Woojin Jun
- Division of Food and Nutrition and Human Ecology Research Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jinhak Kim
- R&D Division, Daehan Chemtech Co. Ltd., Gwacheon 13840, Korea
| | - Kwang-Soo Baek
- R&D Division, Daehan Chemtech Co. Ltd., Gwacheon 13840, Korea
| | - Ok-Kyung Kim
- Division of Food and Nutrition and Human Ecology Research Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jeongmin Lee
- Department of Food Innovation and Health, Kyung Hee University, Yongin 17104, Korea
| |
Collapse
|
28
|
Horton JR, Yu M, Zhou J, Tran M, Anakal RR, Lu Y, Blumenthal RM, Zhang X, Huang Y, Zhang X, Cheng X. Multimeric transcription factor BCL11A utilizes two zinc-finger tandem arrays to bind clustered short sequence motifs. Nat Commun 2025; 16:3672. [PMID: 40246927 PMCID: PMC12006351 DOI: 10.1038/s41467-025-58998-7] [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: 11/01/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BCL11A, a transcription factor, is vital for hematopoiesis, including B and T cell maturation and the fetal-to-adult hemoglobin switch. Mutations in BCL11A are linked to neurodevelopmental disorders. BCL11A contains two DNA-binding zinc-finger arrays, low-affinity ZF2-3 and high-affinity ZF4-6, separated by a 300-amino-acid linker. ZF2-3 and ZF4-5 share 73% identity, including five out of six DNA base-interacting residues. These arrays bind similar short sequence motifs in clusters, with the linker enabling a broader binding span. Crystallographic structures of ZF4-6, in complex with oligonucleotides from the β-globin locus region, reveal DNA sequence recognition by residues Asn756 (ZF4), Lys784 and Arg787 (ZF5). A Lys784-to-Thr mutation, linked to a neurodevelopmental disorder with persistent fetal globin expression, reduces DNA binding over 10-fold but gains interaction with a variable base pair. BCL11A isoforms may form oligomers, enhancing chromatin occupancy and repressor functions by allowing multiple copies of both low- and high-affinity ZF arrays to bind DNA. These distinctive properties, apparently conserved among vertebrates, provide essential functional flexibility to this crucial regulator.
Collapse
Affiliation(s)
- John R Horton
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Meigen Yu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jujun Zhou
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Melody Tran
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rithvi R Anakal
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Robert M Blumenthal
- Department of Medical Microbiology and Immunology, and Program in Bioinformatics, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Xiaotian Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Xing Zhang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Liu A, Bai P, You H, Zhuang Z, Tian F, Weng H, Wei X, Tang L, Wang L, Liu C, Zhang J, Sun M, Zhang S, Shu X, Ge J. SLAMF7 Restrains Pro-Inflammatory Macrophage Activation to Counteract Doxorubicin-Induced Cardiotoxicity. JACC Basic Transl Sci 2025:S2452-302X(25)00073-7. [PMID: 40372307 DOI: 10.1016/j.jacbts.2025.02.015] [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: 12/23/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 05/16/2025]
Abstract
Doxorubicin-induced cardiotoxicity (DIC) poses a significant challenge in cancer treatment. This study investigated the role of SLAMF7 in DIC, particularly in macrophage-mediated inflammation. Using SLAMF7 knockout mice, we found that SLAMF7 deficiency exacerbates DIC and amplifies inflammatory responses. Mechanistically, SLAMF7 interacts with TNF receptor-associated factor 6 to attenuate nuclear factor κB signaling, reducing oxidative stress and proinflammatory cytokines. Notably, administering recombinant SLAMF7 protein effectively mitigated DIC. These findings underscore the critical role of SLAMF7 in protecting against DIC, positioning it as a promising therapeutic target.
Collapse
Affiliation(s)
- Ao Liu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hongmin You
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zehao Zhuang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Fangyan Tian
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Haobo Weng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Xuemei Wei
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Lu Tang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Litao Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaobao Liu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinghong Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Minmin Sun
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Shuning Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xianhong Shu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
30
|
Chen MX, Chen Y, Fu R, Wang JY, Liu SY, Shen TB. Cytomegalovirus infection initiates inflammatory bowel disease by activating a positive MyD88/NF-κB feedback loop in allogeneic skin transplantation mice. Virol J 2025; 22:101. [PMID: 40241182 PMCID: PMC12001603 DOI: 10.1186/s12985-025-02725-7] [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: 09/29/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Infection with the cytomegalovirus (CMV) is common. Inflammatory bowel disease (IBD) is characterized by chronic inflammation in the gastrointestinal tract. CMV infection is involved in IBD pathogenesis. The abnormal activation of myeloid differentiation factor 88 (MyD88)/nuclear factor- kappa B (NF-κB) signaling, which results in inflammatory cytokine overexpression, is an important factor in IBD pathogenesis. The present study aimed to examine the effect of CMV infection on NF-κB activation and its role in IBD pathogenesis. Since BALB/c rather than C57BL/6 mice belong to the murine CMV (MCMV) susceptible strain, allogeneic skin transplantation was conducted between MyD88 (+/+) or MyD88-knockout Myd88 (-/-) BALB/c (recipient) mice and C57BL/6 (donor) mice. Thereafter, the immune function of the recipient mice was reduced by immunosuppressant cyclosporine, which is meaningful in the pathogenesis of IBD caused by MCMV in immunocompromised mice. MCMV strain K181-eGFP (eGFP K181) or hMIEP-eGFP K181 (knockout MCMV IE1-3 promoter) was used to infect MyD88 (+/+) BALB/c mice while eGFP K181 was also used to infect MyD88 (-/-) BALB/c mice on day 14 post allogeneic skin transplantation. MCMV DNA was detected via nested polymerase chain reaction. Hematoxylin-Eosin staining was used to assess colon necrosis and inflammatory cell infiltration. The serum levels of tumor necrosis factor-alpha, interleukin (IL)-1β, IL-6, IL-8, IL-12, flagellin, lipopolysaccharide, and myeloperoxidase were detected by ELISA and immune reaction. Immunoblots were applied to measure protein levels. eGFP K181 infection significantly induced colon permeability, necrosis, inflammatory cell infiltration, and inflammation in allogeneic skin transplantation mice. hMIEP-eGFP K181 infection significantly inhibited colon permeability, necrosis, inflammatory cell infiltration, and inflammation compared with eGFP K181 infection in allogeneic skin transplantation mice. Moreover, the MyD88-dependent NF-κB signaling pathway was involved in the pathogenesis of eGFP K181-induced colon permeability and inflammation in allogeneic skin transplantation mice. Our findings highlight the importance of CMV infection and the Myd88/NF-κB signaling pathway in IBD and might provide a new direction for the development of drugs for IBD.
Collapse
Affiliation(s)
- Ming-Xian Chen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui Road, Hangzhou, 310012, China
- Institute of Integrated Chinese and Western Medicine on Spleen-Stomach Diseases, Zhejiang Province Academy of Traditional Chinese Medicine, Hangzhou, 310012, China
| | - Yu Chen
- Laboratory Animal Center, Zhejiang Province Academy of Traditional Chinese Medicine, Hangzhou, 310007, China
| | - Rui Fu
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui Road, Hangzhou, 310012, China
| | - Jie-Yi Wang
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui Road, Hangzhou, 310012, China
| | - Sai-Yue Liu
- Department of Adverse Drug Reaction Monitoring, Zhejiang Province Center of Adverse Drug Reaction Monitoring, No. 39, Yile Road, Mr, Hangzhou, 310012, China.
| | - Tang-Biao Shen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, No. 234, Gucui Road, Hangzhou, 310012, China.
| |
Collapse
|
31
|
Kogias SS, O'Brien JA, Robertson RV, Peng A, Tinoco-Mendoza FA, Ramachandran A, Henderson LA, Austin PJ. 10-kHz High-Frequency Spinal Cord Stimulation Significantly Reduces Proinflammatory Cytokines and Distinct Populations of T Lymphocytes in Patients With Persistent Spinal Pain Syndrome Type 2. Neuromodulation 2025:S1094-7159(25)00056-X. [PMID: 40232209 DOI: 10.1016/j.neurom.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
OBJECTIVES Persistent spinal pain syndrome type 2 (PSPS-T2) is a chronic mixed nociceptive and neuropathic pain condition that results after lumbar spinal surgery. PSPS-T2 is a highly treatment-resistant condition with less than half of patients receiving adequate pain relief from conventional medications. 10-kHz high-frequency (HF) spinal cord stimulation (SCS) is a highly effective therapy for treatment-resistant PSPS-T2 that can often provide a >50% reduction in pain. This study aimed to systematically investigate the peripheral immune environment in PSPS-T2 compared with that in healthy controls, before assessing the immune effects of 10-kHZ SCS in PSPS-T2. MATERIALS AND METHODS This study used high-parameter mass cytometry and a multiplex cytokine assay to characterize the peripheral immune environment in healthy controls (n = 16) compared with patients with PSPS-T2 before (n = 16) and after seven to ten days of HF SCS treatment (n = 12). RESULTS Compared with healthy controls, there was a significant increase in proinflammatory signaling through nuclear factor-κB, p38 mitogen-activated protein kinase, and signal transducer and activator of transcription 3 pathways in natural killer (NK), CD4+ "terminally differentiated effector memory cells re-expressing CD45RA" (TEMRA), central memory CD8+, and "effector-like" CD8+ T lymphocyte populations in PSPS-T2. Seven to ten days of HF SCS treatment led to significant pain relief in 75% of patients with PSPS-T2, improved psychologic measures, and induced multiple antiinflammatory effects, including a reduction in the abundance of central memory CD4+ T helper 17 (TH17) lymphocytes and natural killer T (NKT) cell populations, that were correlated with pain relief. Furthermore, the expression of granzyme B, a major cytotoxic effector molecule, was reduced in the CD8+ T lymphocyte compartment. These changes in immune cell number and function were associated with a significant reduction in plasma levels of interleukin (IL)-1β, IL-6, IL-8, IL-10, IL-12, interferon (IFN)-α, IFN-γ, and IFN-λ1, and a greater reduction in tumor necrosis factor-α plasma levels in those patients with greatest pain relief, representing an antiinflammatory shift. CONCLUSIONS These changes suggest that PSPS-T2 is a chronic inflammatory condition characterized by cytotoxic and exhausted immune cell populations. The resolution of this inflammation by distinct immune cell populations induced by SCS may contribute to pain relief, and specific populations, such as TH17 and NKT cells, may represent useful biomarkers of treatment effectiveness.
Collapse
Affiliation(s)
- Stefanie S Kogias
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Jayden A O'Brien
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Rebecca V Robertson
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Allan Peng
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Fernando A Tinoco-Mendoza
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Alister Ramachandran
- Pain Management Centre, Westmead Hospital, Westmead, New South Wales, Australia; Pain Med, Norwest, Sydney, New South Wales, Australia
| | - Luke A Henderson
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Paul J Austin
- Brain and Mind Centre, Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.
| |
Collapse
|
32
|
Jeong JS, Ju H, Cho CH. Analysis of the Relationship Between NF-κB1 and Cytokine Gene Expression in Hematological Malignancy: Leveraging Explained Artificial Intelligence and Machine Learning for Small Dataset Insights. Int J Med Sci 2025; 22:2208-2226. [PMID: 40303498 PMCID: PMC12035828 DOI: 10.7150/ijms.109493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
This study measures expression of nuclear factor kappa B (NF-κB)1 and related cytokine genes in bone marrow mononuclear cells in patients with hematological malignancies, analyzing the relationship between them with an integrated framework of statistical analyses, machine learning (ML), and explainable artificial intelligence (XAI). While traditional dimensionality reduction techniques-such as principal component analysis, linear discriminant analysis, and t-distributed stochastic neighbor embedding-showed limited differentiation embedding, ML classifiers (k-Nearest Neighbors, Naïve Bayes Classifier, Random Forest, and XGBoost) successfully identified critical patterns. Notably, normalized caspase-1 counts consistently emerged as the most influential feature associated with NF-κB1 activity across disease groups, as highlighted by SHapley Additive exPlanations analyses. Systematic evaluation of ML performance on small datasets revealed that a minimum sample size of 15-24 is necessary for reliable classification outcomes, particularly in cohorts of acute myeloid leukemia and myelodysplastic syndrome. These findings underscore the pivotal role of caspase-1 to the NF-κB1 gene expression in hematologic malignancy diseases. Furthermore, this study demonstrates the feasibility of leveraging ML and XAI to derive meaningful insights from limited data, offering a robust strategy for biomarker discovery and precision medicine in rare hematological malignancies.
Collapse
Affiliation(s)
- Jae-Seung Jeong
- Division of Artificial Intelligence Convergence Engineering, Sahmyook University, 01795, Republic of Korea
| | - Hyunsu Ju
- Post-Silicon Semiconductor Institute, Korea Institute of Science and Technology, 02792, Republic of Korea
| | - Chi-Hyun Cho
- Department of Laboratory Medicine, College of Medicine, Korea University Ansan Hospital, 15355, Republic of Korea
| |
Collapse
|
33
|
Mohan M, Mannan A, Singh TG. Unravelling the role of protein kinase R (PKR) in neurodegenerative disease: a review. Mol Biol Rep 2025; 52:377. [PMID: 40205152 DOI: 10.1007/s11033-025-10484-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Protein Kinase R is an essential regulator of many cell activities and belongs to one of the largest and most functionally complex gene families. These are found all over the body, and by adding phosphate groups to the substrate proteins, they regulate their activity and coordinate the action of almost all cellular processes. Recent research has illuminated the involvement of PKR in the pathogenesis of neurodegenerative disorders (NDs), thereby expanding our understanding of intricate molecular mechanisms underlying disease progression. Through their inhibition or activation, they hold potential therapeutic targets for the pathogenesis or protection of NDs. In the case of AD (AD), PKR contributes to the protection or elevation of Aβ accumulation, neuroinflammation, synaptic plasticity alterations, and neuronal excitability. Similarly, in Parkinson's disease (PD), PKR again has a dual role in dopaminergic neuronal loss, gene mutations, and mitochondrial dysfunction via various pathways. Notably, neuronal excitotoxicity, as well as genetic mutations, have been linked to ALS. In Huntington's disease (HD), PKR is associated with decreased or increased mutated genes, striatal neuron degeneration, neuroinflammation, and excitotoxicity. This review emphasizes strategies that target PKR for the treatment of neurodegenerative disorders. Doing so offers valuable insights that can guide future research endeavors and the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, Rajpura, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, Rajpura, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, Rajpura, India.
| |
Collapse
|
34
|
Panza P, Kim HT, Lautenschläger T, Piesker J, Günther S, Alayoubi Y, Cleaver O, Looso M, Stainier DYR. The lung microvasculature promotes alveolar type 2 cell differentiation via secreted SPARCL1. Stem Cell Reports 2025; 20:102451. [PMID: 40118055 PMCID: PMC12069885 DOI: 10.1016/j.stemcr.2025.102451] [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: 05/21/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025] Open
Abstract
Lung endothelial cells (ECs) and pericytes are closely juxtaposed with the respiratory epithelium before birth and thus may have instructive roles during development. To test this hypothesis, we screened EC-secreted proteins for their ability to alter cell differentiation in alveolar organoids. We identified secreted protein acidic and rich in cysteine-like protein 1 (SPARCL1) as an extracellular matrix molecule that can promote alveolar type 2 (AT2) cell differentiation in vitro. SPARCL1-treated organoids display lysozyme upregulation and a doubling in the number of AT2 cells at the expense of intermediate progenitors. SPARCL1 also induces the upregulation of nuclear factor κB (NF-κB) target genes, and suppression of NF-κB activation in lung organoids blocked SPARCL1 effects. NF-κB activation by lipopolysaccharide (LPS) was sufficient to induce AT2 cell differentiation; however, pharmacological inhibition of the pathway alone did not prevent it. These data support a role for SPARCL1 and NF-κB in alveolar cell differentiation and suggest a potential value in targeting this signaling axis to promote alveolar maturation and regeneration.
Collapse
Affiliation(s)
- Paolo Panza
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Justus-Liebig University Giessen, Giessen, Germany; Member of the German Center for Lung Research, DZL-UGMLC; Member of the Excellence Cluster Cardio-Pulmonary Institute, CPI.
| | - Hyun-Taek Kim
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Till Lautenschläger
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yousef Alayoubi
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Mario Looso
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Member of the German Center for Lung Research, DZL-UGMLC; Member of the Excellence Cluster Cardio-Pulmonary Institute, CPI.
| |
Collapse
|
35
|
Kennedy AE, Barczewski AH, Arnoldy CR, Pennington JP, Tiernan KA, Hidalgo MB, Reilly CC, Wongsri T, Ragusa MJ, Grigoryan G, Mierke DF, Pellegrini M. The structure of a NEMO construct engineered for screening reveals novel determinants of inhibition. Structure 2025; 33:691-704.e6. [PMID: 39909030 PMCID: PMC11972163 DOI: 10.1016/j.str.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/04/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025]
Abstract
NEMO is an essential component in the activation of the canonical nuclear factor κB (NF-κB) pathway and exerts its function by recruiting the IκB kinases (IKK) to the IKK complex. Inhibition of the NEMO/IKKs interaction is an attractive therapeutic paradigm for diseases related to NF-κB mis-regulation, but a difficult endeavor because of the extensive protein-protein interface. Here we report the design and characterization of novel engineered constructs of the IKK-binding domain of NEMO, programmed to render this difficult protein domain amenable to NMR measurements and crystallization, while preserving its biological function. ZipNEMO binds IKKβ with nanomolar affinity, is amenable to heteronuclear nuclear magnetic resonance (NMR) techniques and structure determination by X-ray crystallography. We show that NMR spectra of zipNEMO allow to detect inhibitor binding in solution and resonance assignment. The crystal structure of zipNEMO reveals a novel ligand binding motif and the adaptability of the binding pocket and inspired the design of new peptide inhibitors.
Collapse
Affiliation(s)
- Amy E Kennedy
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | | | | | | | - Kelly A Tiernan
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | | | | | - Tanyawan Wongsri
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Michael J Ragusa
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Gevorg Grigoryan
- Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA
| | - Dale F Mierke
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Maria Pellegrini
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA.
| |
Collapse
|
36
|
Kewalramani N, Machahua C, Marti TM, Zandbergen C, Chortarea S, Beretta-Piccoli J, von Garnier C, Dorn P, Fytianos K, Funke-Chambour M. Heme-induced lung injury in human precision cut lung slices: a new model for acute lung injury. Respir Res 2025; 26:124. [PMID: 40176049 PMCID: PMC11966866 DOI: 10.1186/s12931-025-03191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) causes high mortality and has no specific pharmacological treatment. Scarcity of drugs against ARDS is in part due to the lack of models for ARDS. As raised serum heme levels are associated with higher mortality in patients with ARDS, we hypothesised that circulating heme contributes to ARDS pathology and can induce lung injury resembling human disease. We aimed to develop a new model for acute lung injury and ARDS research with heme-induced injury in human precision cut lung slices (PCLS). METHODS We analysed heme and its degrading enzymes along with inflammatory cytokines in patients with coronavirus disease 2019 (COVID-19) and ARDS compared to healthy adult subjects. In PCLS, we studied effects of heme on cell survival, membrane integrity, the transcriptome by gene expression and the proteome by protein expression analysis or ELISA. We also tested synergistical effects with lipopolysaccharide (LPS) on cell survival in addition to heme to simulate bacterial infection. RESULTS Patients with COVID-19 and ARDS had increased serum levels of heme and heme oxygenase 1 (HO-1) compared to controls. In PCLS, heme induced cell death in a dose-dependent manner, stimulated pro-inflammatory and injury signals and triggered changes to the extracellular matrix (ECM). Comparative analyses of the lung transcriptomic and proteomic signatures revealed 27 common markers (log2 fold change greater than 1, at adjusted (adj) p-value < 0.05 significant), most of which were inflammatory. Similar inflammatory cytokines were raised in blood from patients with COVID-19 and ARDS compared to controls. LPS did not increase cytotoxicity in addition to heme. CONCLUSION Heme induced inflammatory cytokine release and cell death in human PCLS, resembling the patterns observed in blood samples from patients with COVID-19 and ARDS. Thus, heme-stimulated PCLS represent a novel ex vivo model for mechanistic studies for acute lung injury and ARDS.
Collapse
Affiliation(s)
- Namrata Kewalramani
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Carlos Machahua
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Thomas Michael Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Cas Zandbergen
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | - Savvina Chortarea
- CSL Behring, Research, CSL Biologics Research Center, Bern, Switzerland
| | | | - Christophe von Garnier
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, 1011, Lausanne, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Manuela Funke-Chambour
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
37
|
Clevenger MH, Wei C, Karami AL, Tsikretsis LE, Carlson DA, Pandolfino JE, Gonsalves N, Winter DR, Whelan KA, Tétreault MP. Esophageal epithelial Ikkβ deletion promotes eosinophilic esophagitis in experimental allergy mouse model. J Allergy Clin Immunol 2025; 155:1276-1289. [PMID: 39724973 PMCID: PMC11972898 DOI: 10.1016/j.jaci.2024.12.1070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/19/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic TH2-associated inflammatory disorder triggered by food allergens, resulting in esophageal dysfunction through edema, fibrosis, and tissue remodeling. The role of epithelial remodeling in EoE pathogenesis is critical but not fully understood. OBJECTIVE We investigated the role of epithelial IKKβ/NF-κB signaling in EoE pathogenesis using a mouse model with conditional Ikkβ knockout in esophageal epithelial cells (IkkβEEC-KO). METHODS EoE was induced in IkkβEEC-KO mice through skin sensitization with MC903/ovalbumin followed by intraesophageal ovalbumin challenge. Histologic and transcriptional analyses were performed to assess EoE features. Single-cell RNA sequencing was used to profile esophageal mucosal cell populations and gene expression changes. RESULTS IkkβEEC-KO/EoE mice exhibited hallmark EoE features, including eosinophil infiltration, intraepithelial eosinophils, microabscesses, basal cell hyperplasia, and lamina propria remodeling. RNA sequencing revealed significant alterations in IKKβ/NF-κB signaling pathways, with decreased expression of RELA and increased expression of IKKβ-negative regulators. Sequencing analyses identified disrupted epithelial differentiation and barrier integrity alongside increased type 2 immune responses and peptidase activity. CONCLUSION Loss of epithelial IKKβ signaling exacerbates EoE pathogenesis, highlighting the critical role of this pathway in maintaining epithelial homeostasis and preventing allergic inflammation. The IkkβEEC-KO/EoE mouse model closely mirrors human EoE, providing a valuable tool for investigating disease mechanisms and therapeutic targets. This model can facilitate the development of strategies to prevent chronic inflammation and tissue remodeling in EoE.
Collapse
Affiliation(s)
- Margarette H Clevenger
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Cenfu Wei
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Adam L Karami
- Department of Cancer & Cellular Biology, Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pa
| | - Lia E Tsikretsis
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Dustin A Carlson
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - John E Pandolfino
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Nirmala Gonsalves
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Deborah R Winter
- Department of Medicine, Rheumatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Kelly A Whelan
- Department of Cancer & Cellular Biology, Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pa
| | - Marie-Pier Tétreault
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
38
|
El-Sayed SF, Mahmoud SM, Samy W, Wahid RM, Talaat A, Seada SG. Vitamin D3 mitigates aspirin-induced gastric injury by modulating gastrokines, E-cadherin, and inhibiting NLRP3 and NF-κB/MMP-9 signaling pathway. Tissue Cell 2025; 93:102724. [PMID: 39823708 DOI: 10.1016/j.tice.2025.102724] [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: 10/04/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND The prevalence of gastric ulcers has grown significantly in the modern era affecting 10 % of global population. Aspirin downregulates gastrokines 1(GKN1) expression in gastric mucosa and GKN1 down-regulation results in gastric cancer. Vitamin D3 (Vit.D3) has anti-inflammatory and antioxidant effects. AIM Study the gastroprotective impact of Vit.D3 following aspirin-induced gastric injury in relation to gastrokines and investigate the possible underlying mechanisms. MATERIALS AND METHODS 24 rats were divided into 4 groups: control, Vit.D3 supplemented normal, aspirin-induced gastric injury, and Vit.D3 supplemented gastric injury groups. Some oxidative stress markers with gene expression of GKN1&2, mucin 5AC (Muc5ac) and NLR family pyrin domain containing 3 (NLRP3) in the gastric tissue were done. Histopathological and immunohistochemical study of E-Cadherin, nuclear factor kappa beta (NFκB), and metalloproteinase-9 (MMP-9) in the stomach mucosa were identified. RESULTS Vit.D3 supplementation significantly upregulated E-Cadherin, GSH, GKN1 and Muc5ac in the gastric tissue. Also, it improved the morphology, histology of gastric tissue, by alleviating oxidative stress and NFκB, MMP-9 and down regulation of inflammasome (NLRP3). CONCLUSION Vitamin D3 has a potential protective effect against aspirin -induced gastric injury via upregulating gastrokine1 and E-cadherin and down regulation of NFKB/MMP-9 signaling pathway and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Sherein F El-Sayed
- Department of Medical Physiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Samar Mortada Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Walaa Samy
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Reham M Wahid
- Department of Medical Physiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Aliaa Talaat
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Sara G Seada
- Department of Medical Physiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
39
|
Zhu W, Zeng S, Zhu S, Zhang Z, Zhao R, Qiu Q, Luo Z, Qin Y, Chen W, Li B, He Y, Yi L, Ding H, Zhao M, Chen J, Fu C, Fan S. Histone H2B lysine lactylation modulates the NF-κB response via KPNA2 during CSFV infection. Int J Biol Macromol 2025; 299:139973. [PMID: 39826749 DOI: 10.1016/j.ijbiomac.2025.139973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/23/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Histone lysine lactylation (Kla) has recently been reported to participate in various biological processes, regulating transcription, inflammation, and immune-related diseases. However, the mechanism of histone Kla in innate immunity and viral infection remains largely unknown. Here, we observed fluorescent Kla signals in all four histones (H2A, H2B, H3, and H4) in PK-15 cells. Immunoprecipitation analysis showed prominent histone Kla protein bands, with H2B being the most abundant. We generated the H2B K16R mutant plasmid and identified K16 as one of the Kla modification sites in H2B. Further exploration revealed increased global H2B Kla and H2BK16la levels upon classical swine fever virus (CSFV) infection. By employing the Kla agonist (L-lactate), inhibitor (oxamate), or siLDHA, we demonstrated that H2BK16la and pan Kla in PK-15 cells rely on the LDHA-lactate axis, which is also crucial for CSFV-induced H2BK16la and pan Kla levels. Moreover, our data proved the interaction between H2B and CSFV NS4A protein. Notably, H2B Kla can modulate CSFV proliferation. Mechanistically, H2BK16la and pan Kla activate the nuclear factor kappa B (NF-κB) pathway by mediating p65 nuclear translocation via karyopherin α2 (KPNA2), thereby inducing type III interferon (IFN-λ) expression and inhibiting CSFV replication. In conclusion, our study unveils the role of H2B Kla in regulating the NF-κB pathway during viral infection, presenting a novel mechanism. These findings significantly contribute to understanding the pathogenic mechanisms during viral infection and hold promise for the development of viral therapeutic strategies.
Collapse
Affiliation(s)
- Wenhui Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Sen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuaiqi Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhanhui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ruibo Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qi Qiu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zipeng Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuwei Qin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Bingke Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yintao He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China.
| | - Cheng Fu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, PR China.
| |
Collapse
|
40
|
Kim D, Allen CA, Chung D, Meng L, Zhang X, Zhang W, Ouyang Y, Li Z, Hong F. A novel TLR4 accessory molecule drives hepatic oncogenesis through tumor-associated macrophages. Cancer Lett 2025; 614:217543. [PMID: 39929433 DOI: 10.1016/j.canlet.2025.217543] [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: 10/24/2024] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment, yet the roles and mechanisms of TAMs in inflammation-associated oncogenesis remain enigmatic. We report that protein canopy homolog 2 (CNPY2) functions as a novel TLR4 regulator, promoting cytokine production in macrophages. CNPY2 binds directly to TLR4. Cnpy2 deficiency reduces cell surface expression of TLR4, nuclear translocation of NFκB and cytokine production in macrophages. Macrophage-specific CNPY2 deficiency significantly decreases cytokine production in macrophages and reduces hepatocarcinogenesis in a diethylnitrosamine (DEN)-induced liver cancer model. RNA-sequencing analysis revealed Cnpy2 knockout decreased the mRNA level and cell surface expression of two VEGF receptors, Flt1 and Kdr, compared to those in WT counterparts, resulting in inhibition of macrophage tumor infiltration. Cnpy2 knockout inhibits NFκB2/p52-mediated transcription of Flt1 and Kdr in macrophages. These findings demonstrate that CNPY2 regulates macrophages in both inflammation and hepatocarcinogenesis and may serve as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Doyeon Kim
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Carter A Allen
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Dongjun Chung
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Biostatistics Core, College of Nursing, College of Public Health, University of South Florida Health, 12901 Bruce B. Downs Blvd.Tampa, FL, 33612, USA
| | - Wenqing Zhang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Yuli Ouyang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Zihai Li
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA
| | - Feng Hong
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
41
|
HAN S, Du Z, WANG Z, HUANG T, GE Y, SHI J, GAO J. Network pharmacology approach to unveiling the mechanism of berberine in the amelioration of morphine tolerance. J TRADIT CHIN MED 2025; 45:376-384. [PMID: 40151124 PMCID: PMC11955763 DOI: 10.19852/j.cnki.jtcm.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/23/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To investigate the mechanism underlying the effect of the Huanglian decoction (, HLD) on morphine tolerance (MT), using network pharmacology, and to verify these mechanisms in vitro and in vivo. METHODS Available biological data on each drug in the HLD were retrieved from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform. The target proteins of MT were retrieved from the GeneCards, PharmGkb, Therapeutic Target Database, DrugBank, and Online Mendelian Inheritance in Man databases. Information regarding MT and the drug targets was compared to obtain overlapping elements. This information was imported into the Search Tool for the Retrieval of Interacting Genes/Proteins platform to obtain a protein-protein interaction network diagram. Then, a "component-target" network diagram was constructed using screened drug components and target information, viaCytoscape (Institute for Systems Biology, Seattle, WA, USA). The database for annotation, visualization, and integrated discovery was used for Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathways analyses. Pathway information predicted by network pharmacology was verified using animal studies and cell experiments. RESULTS Network pharmacology analysis identified 22 active compounds of HLD and revealed that HLD partially ameliorated MT by modulating inflammatory, apoptosis, and nuclear factor kappa B (NF-κB) signaling pathways. Berberine (BBR), one of the main components of HLD, inhibited the development of MT in mice. BBR reduced cell viability while increasing B-cell lymphoma 2 (Bcl-2) protein expression and decreasing CD86, NF-κB, Bax, and Caspase-3 protein expression in brain vascular 2 (BV2) mcroglia cells treated with morphine. Additionally, BBR contributed to a reduction in pro-inflammatory cytokine release and apoptotic cell number. CONCLUSIONS BBR, a key component of HLD, effectively suppressed microglial activation and neuro-inflammation by regulating the NF-κB and apoptosis signaling pathways, thereby delaying MT. This study offers a novel approach to enhance the clinical analgesic efficacy of morphine.
Collapse
Affiliation(s)
- Shuai HAN
- 1 Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- 2 Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
- 3 Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225009, China
| | - Zhikang Du
- 1 Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- 2 Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Zirui WANG
- 1 Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- 2 Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Tianfeng HUANG
- 3 Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225009, China
| | - Yali GE
- 3 Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225009, China
| | - Jianwen SHI
- 4 Peking University People's Hospital, Qingdao 266111, China
- 5 Women and Children's Hospital, Qingdao University, Qingdao 266034, China
| | - Ju GAO
- 3 Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
42
|
Tao W, Jiang C, Velu P, Lv C, Niu Y. Rosmanol Suppresses Nasopharyngeal Carcinoma Cell Proliferation and Enhances Apoptosis, the Regulation of MAPK/NF-κB Signaling Pathway. Biotechnol Appl Biochem 2025:e2750. [PMID: 40170441 DOI: 10.1002/bab.2750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/08/2025] [Indexed: 04/03/2025]
Abstract
Nasopharyngeal Carcinoma (NPC) is a major public health problem in endemic zones. NPC is correlated with substantial illness and death; thus, superior treatment is desired. Rosmanol (RM) is a phenolic diterpene antioxidant extracted from the medicinal herb Rosemary (Rosmarinus officinalis). RM has been investigated for its anti-inflammatory and anti-tumor properties by numerous signaling cascades. However, the fundamental anticancer latent mechanism of RM persists as unidentified. Hence, this present research proposes to search for the anti-cancer efficacy of RM on human NPC cells CNE2 using an in vitro approach. To assess the possible molecular mechanisms of proliferation, apoptosis, cell-cycle regulatory proteins, and MAPKs/NF-κB signaling of NPC cells were administered RM (20 and 30 µM) and assayed through MTT, DCFH-DA, Rh-123 staining, AO/EB, PI, Rh-123/DAPI merge form staining, RT-PCR, and Western blot. The result was recognized that RM could reduce NPC cell viability by elevated intracellular ROS, MMP damage, and generate apoptosis. RM inhibits the Cyclin-D1, Bax, TNF-α, and NF-κB, and induces BCl-2 analyzed via RT PCR. RM attenuates the cell cycle mechanism by repressing NPC cell cycle-related proteins: CDK4/CDK6, pRB, cyclin-D1, and MAPKs/NF-κB signaling. These data established that the MAPKs/NF-κB pathway is a potential target for the remedial action of RM. In summary, RM may be an effective conventional chemotherapy drug in preventing the progression of NPC.
Collapse
Affiliation(s)
- Weiping Tao
- Department of Otorhinolaryngology-Head and Neck Surgery, The Fourth Hospital of Changsha, Changsha, China
| | - Chaowu Jiang
- Department I of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming City, China
| | - Periyannan Velu
- Galileovasan Offshore and Research and Development Pvt Ltd, Nagapattinam, Tamil Nadu, India
| | - Cao Lv
- Department of Otolaryngology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yan Niu
- Department of Otolaryngology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
43
|
Marín V, Villegas C, Ogundele AV, Cabrera-Pardo JR, Schmidt B, Paz C, Burgos V. Inhibitory Potential of the Drimane Sesquiterpenoids Isotadeonal and Polygodial in the NF-kB Pathway. Molecules 2025; 30:1555. [PMID: 40286191 PMCID: PMC11990674 DOI: 10.3390/molecules30071555] [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: 02/12/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammation contributes to the onset and development of many diseases, including neurodegenerative diseases, caused by the activation of microglia, leading to neurological deterioration. Nuclear factor-κB (NF-κB) is one of the most relevant pathways for identifying anti-inflammatory molecules. In this study, polygodial and isotadeonal, two drimane sesquiterpene dialdehydes, were isolated from Drimys winteri, a medicinal tree of the Mapuche people in Chile. Isotadeonal, or epi-polygodial, was obtained from polygodial by epimerization in basic media (60% yield, Na2CO3, r/t, 24 h). Both sesquiterpenoids were evaluated on the NF-κB pathway, with the result that isotadeonal inhibited the phosphorylation of IκB-α at 10 μM with higher potency by Western blotting. The final inhibition of the pathway was evaluated using a SEAP reporter (secreted alkaline phosphatase) on THP-1 cells. Isotadeonal inhibited SEAP with higher potency than polygodial, quercetin, and CAPE (phenethyl ester of caffeic acid). In silico analysis suggests that the α-aldehyde of isotadeonal adopts a more stable conformation in the active pocket of IκB-α than polygodial.
Collapse
Affiliation(s)
- Víctor Marín
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (V.M.); (A.V.O.)
| | - Cecilia Villegas
- Departamento de Ciencias Biológicas y Químicas, Facultad de Recursos Naturales, Universidad Católica de Temuco, Rudecindo Ortega, Temuco 4780000, Chile;
| | - Ayorinde Víctor Ogundele
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (V.M.); (A.V.O.)
| | - Jaime R. Cabrera-Pardo
- Laboratorio de Química Aplicada y Sustentable (LabQAS), Departamento de Química, Universidad del Bío-Bío, Avenida Collao 1202, Concepcion 4051381, Chile;
- College of Dental Medicine, Roseman University of Health Sciences, 10894 S. River Front Parkway, South Jordan, UT 84095, USA
| | - Bernd Schmidt
- Institut für Chemie, Universität Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany;
| | - Cristian Paz
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (V.M.); (A.V.O.)
| | - Viviana Burgos
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Temuco 4780000, Chile
| |
Collapse
|
44
|
Mellgren AEC, Cristea I, Stevenson T, Spriet E, Knappskog PM, Bøe SO, Kranz H, Grellscheid SN, Rødahl E. On subcellular distribution of the zinc finger 469 protein (ZNF469) and observed discrepancy in the localization of endogenous and overexpressed ZNF469. FEBS Open Bio 2025. [PMID: 40156465 DOI: 10.1002/2211-5463.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025] Open
Abstract
The zinc finger 469 gene (ZNF469) is a single-exon gene predicted to encode a protein of 3953 amino acids. Despite pathogenic ZNF469 variants being associated with Brittle Cornea Syndrome (BCS), relatively little is known about ZNF469 beyond its participation in regulating the expression of genes encoding extracellular matrix proteins. In this study, we examined the expression and intracellular localization of ZNF469 in different cell lines. The level of ZNF469 mRNA varied from low levels in HEK293 cells to high levels in HeLa cells and primary fibroblasts. Antibodies against ZNF469 reacted among others with a protein of approximately 400 kDa in immunoblot analysis, which was mainly present in the insoluble fraction of the cytoplasm. Immunofluorescence analysis of interphase cells showed small cytoplasmic puncta and weak nuclear staining. In dividing HeLa cells, the antibodies recognized foci that also stained for proteasomes. In transfected cells, ZNF469 was observed mainly in foci resembling nuclear speckles in interphase and at the midbody during mitosis. The nuclear foci showed overlapping staining with proteasomes. In live cell imaging, liquid-like properties of the nuclear foci were recorded as they changed shape and position and occasionally fused with each other. During stress granule formation, cytoplasmic foci showed overlapping staining with G3BP1. Finally, in silico analysis revealed large intrinsically disordered regions with multiple low complexity domains in ZNF469. Our data indicate that ZNF469 forms aggregates possibly as biomolecular condensates when overexpressed. However, care must be taken when analyzing the intracellular distribution of ZNF469 due to the discrepancy in the localization of endogenous ZNF469 and overexpressed ZNF469 in transfected cells.
Collapse
Affiliation(s)
| | - Ileana Cristea
- Department of Clinical Medicine, University of Bergen, Norway
- Department of Ophthalmology, Haukeland University Hospital, Norway
| | - Thomas Stevenson
- Computational Biology Unit and Department of Biomedicine, University of Bergen, Norway
| | - Endy Spriet
- Molecular Imaging Center, Department of Biomedicine, University of Bergen, Norway
| | - Per Morten Knappskog
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Norway
| | - Stig Ove Bøe
- Department of Microbiology, Oslo University Hospital, Norway
| | - Harald Kranz
- Gen-H Genetic Engineering Heidelberg GmbH, Heidelberg, Germany
| | - Sushma N Grellscheid
- Computational Biology Unit and Department of Biomedicine, University of Bergen, Norway
| | - Eyvind Rødahl
- Department of Clinical Medicine, University of Bergen, Norway
- Department of Ophthalmology, Haukeland University Hospital, Norway
| |
Collapse
|
45
|
Yang B, Cao HX, He YW, Ouyang JJ, Lv M, Li YX, Lu YD. Increased keratin 80 expression predicts poor prognosis and promotes oxaliplatin resistance in gastric cancer. World J Gastroenterol 2025; 31:103991. [PMID: 40182597 PMCID: PMC11962845 DOI: 10.3748/wjg.v31.i12.103991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/26/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Keratin 80 (KRT80), a type I intermediate filament protein, is a member of the keratin family with specialized functions in epithelial tissues. While KRT80 has been implicated in both normal physiological processes and various diseases, its role in gastric cancer (GC), particularly its expression and prognostic significance, remains poorly understood. In this study, we investigated the role and underlying molecular mechanisms of KRT80 in oxaliplatin resistance in GC. Our analysis revealed that KRT80 is significantly upregulated in GC tissues and is associated with poor clinical prognosis. The role of KRT80 in GC cell proliferation was assessed through in vitro and in vivo assays. AIM To explore the expression of KRT80 in GC and its impact on the prognosis of patients. METHODS KRT80 expression in GC tissues was analyzed using Western blotting, quantitative reverse transcription PCR, multiple immunofluorescence staining, and immunohistochemistry. Survival analysis was conducted using the Kaplan-Meier method with the log-rank test. The role of KRT80 in GC cell proliferation was assessed through in vitro and in vivo assays. Immunoprecipitation and mass spectrometry analyses identified elongation factor 1-alpha 1 (EEF1A1) as a binding protein of KRT80. RESULTS Integrating our experimental findings with multiple published studies, we found that increased KRT80 expression is associated with poor prognosis in GC and promotes resistance to oxaliplatin. Moreover, we have preliminarily verified the interaction between KRT80 and EEF1A1. Therefore, this study provides a novel perspective on overcoming oxaliplatin resistance in GC. CONCLUSION Increased KRT80 expression predicts poor prognosis and promotes oxaliplatin resistance in GC, suggesting its potential as a novel prognostic biomarker.
Collapse
Affiliation(s)
- Bo Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Hong-Xia Cao
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Ya-Wei He
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Ji-Jie Ouyang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Meng Lv
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Yong-Xiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Yi-Da Lu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| |
Collapse
|
46
|
Yang Y, Du Y, Ma X, Yuan G, Li G, Zhang Q, Zhou S. Transcription factor addictions: exploring the potential Achilles' Heel of endometriosis. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2832-8. [PMID: 40163264 DOI: 10.1007/s11427-024-2832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/15/2024] [Indexed: 04/02/2025]
Abstract
A considerable number of women of reproductive age suffer from endometriosis worldwide. There is a significant physical, mental, and financial burden on patients affected by this condition in terms of pelvic pain, either continuously or intermittently, dysmenorrhea, infertility, and a higher risk of certain types of cancer. Several treatments available in clinical settings for endometriosis management do not provide adequate efficacy and have undesirable side effects. Transcription factors (TFs) are crucial regulators of key biological processes involved in endometriosis. Here, we elaborated on the research progress regarding the crucial roles of TFs in endometriosis, emphasizing their implications for clinical outcomes and critical therapeutic contributions. By delving into their involvement in key processes, such as cell proliferation and apoptosis, we revealed the multifaceted role of key TFs in disease progression. We aimed to provide a systemic understanding of TFs regulation in endometriosis pathogenesis, establishing a foundation for innovative treatment approaches.
Collapse
Affiliation(s)
- Yang Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Yi Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Yuan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Guobo Li
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
47
|
Manikandan C, Jaiswal AK. Therapeutic Potential of NF-κB Inhibition in Glioblastoma: Gene Therapy Approach with rAAV-5 Mediated IκBαM Overexpression. Mol Biotechnol 2025:10.1007/s12033-025-01418-4. [PMID: 40140181 DOI: 10.1007/s12033-025-01418-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/21/2025] [Indexed: 03/28/2025]
Abstract
Current treatment strategies for glioblastoma, including resection followed by concurrent chemotherapy/radiotherapy are not curative. Angiogenesis and hypoxia are two major factors responsible for GBM growth and resistance to existing therapies, leading to poor clinical outcomes. The transcription factor NF-κB induces tumour progression by activating genes associated with cell proliferation and angiogenesis. It is expressed constitutively in gliomas and is known to regulate the expression of HIF-1α and VEGF in GBM. As a result, NF-κB can be a potent target that can inhibit tumour growth/invasiveness by reducing hypoxia and angiogenesis, as well as preventing macrophage and microglia infiltration and generating inflammatory cytokines that cause gliomagenesis. AAV vectors are the typical transducing agents for gene therapy because they can infect a broad range of dividing and non-dividing cell types. AAVs have emerged as one of the most widely used methods for delivering genes into the central nervous system because of their broad range of infectivity, ability to induce long-term transgenic expression, and lack of toxicity. The present study aims to inhibit NF-κB activity by blocking its nuclear translocation via overexpression of IκBα utilising recombinant adeno-associated virus-5 plasmid as a gene therapy vector.
Collapse
Affiliation(s)
- Ceera Manikandan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, 632014, India
| | - Amit Kumar Jaiswal
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
48
|
Mongi-Bragato B, Sánchez MA, Avalos MP, Boezio MJ, Guzman AS, Rigoni D, Perassi EM, Mas CR, Bisbal M, Bollati FA, Cancela LM. Activation of Nuclear Factor-kappa B in the nucleus accumbens core is necessary for chronic stress-induced glutamate and neuro-immune alterations that facilitate cocaine self-administration. Brain Behav Immun 2025; 128:1-15. [PMID: 40139275 DOI: 10.1016/j.bbi.2025.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Stressful events are associated with impaired glutamate signaling and neuroimmune adaptations that may increase the vulnerability of individuals to cocaine addiction. We previously demonstrated that chronic stress induced reactive microglia and increased TNF-α expression in the nucleus accumbens core (NAcore), both alterations strongly linked with impaired glutamate homeostasis and the facilitation of cocaine self-administration. The nuclear factor kappa-B (NF-κB) is a critical regulator of many immune- and addiction-related genes, such as the gene coding for glutamate transporter (GLT-1), and it is considered a master regulator of inflammation, reported to be a key driver of microglia activation in psychiatric diseases. However, no studies have examined the role of NF-κB signaling within the NAcore in the neuroimmune and glutamate mechanism, underpinning stress-induced vulnerability to cocaine self-administration. Here we investigate whether viral dominant negative inhibition of I kappa B kinase (IKKdn), a signaling molecule responsible for NF-κB activation, would prevent stress-induced facilitation to cocaine self-administration and associated changes in accumbal GLT-1 and TNF-α expression. We also explore N-myc proto-oncogene protein (N-myc) levels as a link between NF-κB and stress-induced GLT-1 downregulation. For seven days (days 1-7), adult male rats were restrained for 2 h/day. Animals were administered an intra-NAcore with IKKdn or empty lentiviruses on day 14 after the first restraint stress session. Marked activation of NF-κB was detected in the NAcore of stressed subjects, along with increased NF-κB expression in astrocytes. Consistently, viral NF-κB inhibition prevented stress-induced facilitation of cocaine self-administration. Moreover, NF-κB blockade results in the restoration of stress-induced reduction in GLT-1 levels and was effective in suppressing stress-induced TNF-α within the NAcore. These findings suggest that accumbal NF-κB signaling exerts a central control over stress-altered downstream neuroimmune and glutamate function underlying vulnerability to cocaine use disorders.
Collapse
Affiliation(s)
- Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| | - Marianela Adela Sánchez
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - María Paula Avalos
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - María Julieta Boezio
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Andrea Susana Guzman
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Diana Rigoni
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Eduardo Marcelo Perassi
- Instituto de Investigaciones en Físico-Química de Córdoba, INFIQC-CONICET, Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Carlos Ruben Mas
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC-CONICET, Departamento de Química Bilógica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Mariano Bisbal
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Friuli 2434, Colinas de Vélez Sarsfield (5016) Córdoba, Argentina, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Flavia Andrea Bollati
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| | - Liliana Marina Cancela
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| |
Collapse
|
49
|
He J, Chen Y, Li Y, Feng Y. Molecular mechanisms and therapeutic interventions in acute kidney injury: a literature review. BMC Nephrol 2025; 26:144. [PMID: 40121405 PMCID: PMC11929251 DOI: 10.1186/s12882-025-04077-4] [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: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical challenge characterized by elevated morbidity and a substantial impact on individual health and socioeconomic factors. A comprehensive examination of the molecular pathways behind AKI is essential for its prevention and management. In recent years, vigorous research in the domain of AKI has concentrated on pathophysiological characteristics, early identification, and therapeutic approaches across many aetiologies and highlighted the principal themes of oxidative stress, inflammatory response, apoptosis, necrosis, and immunological response. This review comprehensively reviewed the molecular mechanisms underlying AKI, including oxidative stress, inflammatory pathways, immune cell-mediated injury, activation of the renin-angiotensin-aldosterone (RAAS) system, mitochondrial damage and autophagy, apoptosis, necrosis, etc. Inflammatory pathways are involved in the injuries in all four structural components of the kidney. We also summarized therapeutic techniques and pharmacological agents associated with the aforementioned molecular pathways. This work aims to clarify the molecular mechanisms of AKI thoroughly, offer novel insights for further investigations of AKI, and facilitate the formulation of efficient therapeutic methods to avert the progression of AKI.
Collapse
Affiliation(s)
- Jiajia He
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanqin Chen
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Li
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China.
| |
Collapse
|
50
|
Hodge N, Tétreault MP. Epithelial Ikkβ deletion modulates immune responses and the IFNγ/CXCL9 axis during early esophageal carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643566. [PMID: 40166246 PMCID: PMC11957055 DOI: 10.1101/2025.03.18.643566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Esophageal cancer is a major cause of cancer-related death, often preceded with chronic inflammation and injuries. The NFκB/IKKβ pathway plays a central role in inflammation, yet its role in early esophageal carcinogenesis remains unclear. This study investigated the role of epithelial IKKβ in early esophageal carcinogenesis. Mice were treated with the carcinogen 4-nitroquinoline-1-oxide (4-NQO) or a vehicle for one month to induce precancerous lesions. Esophagi were harvested and examined through histological, protein, flow cytometry, and RNA analyses. Histological analysis revealed that 4-NQO treatment led to increased inflammation, intraepithelial CD45+ immune cells, and elevated IKKβ phosphorylation levels. Mice with esophageal epithelial-specific Ikkβ deletion (4-NQO/Ikkβ EEC-KO ) showed delayed progression to a precancerous state, with reduced immune cell recruitment compared to 4-NQO/controls. Immunophenotyping showed decreased recruitment of T cells, including CD4+, CD8+ and regulatory (Tregs) T cells, and increased recruitment of macrophages in 4-NQO/Ikkβ EEC-KO mice compared to 4-NQO/controls. RNA sequencing data identified 262 differentially expressed genes in 4-NQO/Ikkβ EEC-KO mice, implicating pathways related to inflammation and wound healing. Notably, the chemokine CXCL9, a T cell chemoattractant, was significantly upregulated in 4-NQO control mice, but not in 4-NQO/Ikkβ EEC-KO mice. Further analysis identified IFNγ as an upstream regulator of Cxcl9 expression, and neutralization of IFNγ reduced Cxcl9 expression levels in 4-NQO treated mice. Additionally, in vitro studies demonstrated that IFNγ upregulates Cxcl9 in an NF-κB dependent manner in esophageal keratinocytes. These findings suggest that epithelial IKKβ regulates the immune microenvironment in early esophageal carcinogenesis through the IFNγ/CXCL9 axis and influencing T cell recruitment and inflammatory responses.
Collapse
Affiliation(s)
- Nathan Hodge
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611-3010, USA
| | - Marie-Pier Tétreault
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611-3010, USA
| |
Collapse
|