1
|
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
|
2
|
Guan Y, Ruan J, Tan P, Qian S, Zhou S, Zhang A, Fu Y, Zhao S, Ran Y, Feng X, Wang Y, Wu X, Zhang B, Ji W, Wu L, Guo X. Hesperidin alleviates endothelial cell inflammation and apoptosis of Kawasaki disease through inhibiting the TLR4/IĸBα/NF-ĸB pathway. Chem Biol Interact 2025; 411:111445. [PMID: 39987982 DOI: 10.1016/j.cbi.2025.111445] [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: 12/19/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Kawasaki Disease (KD) is an acute and self-limiting vasculitis of unknown etiology that mainly occurs in infancy and can lead to vascular endothelial injury. Hesperidin (HES) is an economical dietary biological flavonoid with anti-oxidant, anti-inflammatory, and anti-apoptotic pharmacological effects. The main objective of this study was to investigate the protective effects of HES on KD, and try to elucidate the underlying mechanism. The Candida albicans water-soluble fraction (CAWS) was used to induce coronary arteritis of KD mouse model in vivo, and tumor necrosis factor α (TNF-α) was employed to induce human umbilical vein endothelial cell (HUVEC) injury of KD cell model in vitro to investigate the anti-inflammatory and anti-apoptotic effects of HES on KD. Our in vivo results showed that HES significantly reduced coronary artery injury in KD mice by alleviating pericoronary inflammatory infiltration and tissue fibrosis, inhibiting inflammatory cytokines and chemokine expressions, and decreasing vascular endothelial cell apoptosis. Our in vitro study confirmed that HES had the opposite ability of the NF-κB agonist NF-ĸB activator 1 (ACT1) to significantly alleviate the inflammatory response, CellROX level, and apoptosis by decreasing BAX/BCL-2 and Cleaved Caspase-3 levels as well as reducing TUNEL positive cells and the ratio of flow cytometric apoptotic cells in TNF-α induced HUVECs. The further mechanism study based on bioinformatics analysis and western blotting demonstrated that HES could protect against vascular inflammation and cell apoptosis of KD through inhibiting the TLR4/IĸBα/NF-ĸB pathway, suggesting that HES may be a promising therapeutic candidate for KD.
Collapse
Affiliation(s)
- Yuting Guan
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinghua Ruan
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Pingping Tan
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songwei Qian
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Size Zhou
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ao Zhang
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuchong Fu
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuhui Zhao
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Ran
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing Feng
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijia Wang
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinlei Wu
- Zhejiang-Ireland Joint Laboratory for Precision Diagnosis and Treatment of Valvular Heart Diseases, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang, China
| | - Bing Zhang
- Engineering Research Center of Techniques and Instruments for Diagnosis and Treatment of Congenital Heart Disease, Institute of Developmental and Regenerative Medicine, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiping Ji
- Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China; Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Lianpin Wu
- Zhejiang-Ireland Joint Laboratory for Precision Diagnosis and Treatment of Valvular Heart Diseases, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang, China.
| | - Xiaoling Guo
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Maurya R, Sharma A, Naqvi S. Decoding NLRP3 Inflammasome Activation in Alzheimer's Disease: A Focus on Receptor Dynamics. Mol Neurobiol 2025:10.1007/s12035-025-04918-1. [PMID: 40232645 DOI: 10.1007/s12035-025-04918-1] [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: 12/02/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) is a leading neurodegenerative disorder marked by progressive cognitive decline and significant neuropsychiatric disturbances. Neuroinflammation, mediated by the NLRP3 inflammasome, is increasingly recognized as a critical factor in AD pathogenesis. The NLRP3 inflammasome, a crucial component of the innate immune system, is activated in response to both pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). In AD, amyloid-beta (Aβ) plaques and tau aggregates act as DAMPs, triggering NLRP3 inflammasome activation in microglia and astrocytes. This activation leads to the production of pro-inflammatory cytokines IL-1β and IL-18, contributing to chronic neuroinflammation and neuronal death. This review explores the intricate mechanisms involved in NLRP3 activation, with a particular focus on TREM-2, Msn Kinase MINK, NF-κB, Toll-like receptors, and P2X7 receptors. Understanding these mechanisms offers insight into the multifaceted regulation of the NLRP3 inflammasome and its impact on AD pathology. By elucidating the roles of TREM-2, MINK1, NF-κB, TLRs, and P2X7 receptors, this review highlights potential therapeutic targets for modulating NLRP3 activity. Targeting these pathways could offer novel strategies for mitigating neuroinflammation and slowing the progression of AD. The interplay between these receptors and signaling pathways underscores the complexity of NLRP3 inflammasome regulation and its significance in AD, providing a foundation for future research aimed at developing effective therapeutic interventions.
Collapse
Affiliation(s)
- Ranika Maurya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India
| | - Abha Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India
| | - Saba Naqvi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India.
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India.
| |
Collapse
|
4
|
Al Shahrani M, AboHassan M, Gahtani R, Alshahrani MY, Suliman M, Ahmad I, Saeed M. High-throughput screening and in vitro evaluation of CSB-0914; a novel small molecule NF-κB inhibitor attenuating inflammatory responses through NF-κB, Nrf2 and HO-1 cross-talk. J Biomol Struct Dyn 2025; 43:2762-2771. [PMID: 38127429 DOI: 10.1080/07391102.2023.2294377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Unpleasant side effects of standard inflammatory drugs urges search for novel therapeutic candidates. This study aims in identifying novel anti-inflammatory NF-κB inhibitor by high-throughput computational and in-vitro pre-clinical approaches. Lead candidate selection was conducted by the use of computational docking molecular-dynamic simulations. The RBL-2H3 cell line, derived from rat basophils, was used to evaluate the release of cytokines and degranulation. The study focused on the study of neutrophil elastase and its role in cellular motility. Flow cytometry was utilized to evaluate the activation of basophils and the expression of critical signaling proteins. High throughput screening identified CSB-0914 to stably bind NF-κB-p50 subunit. Dose based loss in T NF-α and IL-2 release were observed in RBL-2H3 cells in addition to degranulation inhibition by CSB-0914. The compound demonstrated significant efficacy in reducing basophil activation assay induced by FcεRI receptors, with an IC50 value of 98.41 nM.. A dose dependent decrease in neutrophil migration and elastase were observed when treated with CSB- 0914. The compound was effective in decreasing. Upon stimulation, RBL-2H3 cells exhibited phosphorylation of NF-κB p-65 as well as upregulation of the Nrf2 and HO-1 signaling pathways. Collectively, our study has successfully identified a novel inhibitor called CSB-0914 that effectively regulates inflammatory responses. These reactions are primarily mediated by the interplay between NF-κB, Nrf2, and HO-1. The findings of this study provide support for the need to conduct more research on CSB-0914 with the aim of its development as a pharmaceutical agent for anti-inflammatory purposes.
Collapse
Affiliation(s)
- Mesfer Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad AboHassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Reem Gahtani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
5
|
Saleh SR, Agwah RG, Elblehi SS, Ghareeb AZ, Ghareeb DA, Maher AM. Combination of 10-hydroxy-decanoic acid and ZnO nanoparticles abrogates lead acetate-induced nephrotoxicity in rats: targeting oxidative stress and inflammatory signalling. BMC Pharmacol Toxicol 2025; 26:69. [PMID: 40134036 PMCID: PMC11934796 DOI: 10.1186/s40360-025-00888-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
Lead is a heavy metal contaminant that can cause significant alterations in renal structure and function, resulting in nephrotoxicity. The fatty acids of royal jelly exhibit immunoregulatory, anticancer, anti-inflammatory, and antioxidant properties, which have garnered significant interest. The most prevalent among them is 10-hydroxydecanoic acid (10-HDA). Zinc oxide nanoparticles (ZnONPs) demonstrate a renoprotective effect, likely due to their antioxidant, anti-inflammatory, and antiapoptotic properties. This study evaluated the therapeutic efficacy of 10-HDA and ZnONPs, administered either as monotherapy or in combination, against lead-induced nephrotoxicity. Male rats were orally administered lead acetate (PbAc) for three months, followed by the administration of 10-HDA and/or ZnONPs for one month. Exposure to PbAc resulted in elevated renal lead concentration, as well as increased serum levels of urea, creatinine, and cystatin C. The condition resulted in damage to the renal parenchyma, characterised by degenerative glomeruli and tubules, and exhibited the highest lesion score. Nrf2 and HO-1 exhibited reduced expression and diminished antioxidant enzyme levels subsequent to PbAc poisoning. Additionally, there was an increase in the inflammatory and apoptotic signalling through the p-IKK/NF-κB axis. The administration of 10-HDA and ZnONPs significantly decreased renal lead levels and improved antioxidant capacity. Moreover, renal inflammatory markers (TNF-α, p-IKK, IL-1β, IL-6, and IL-8) and proapoptotic indicators (Bax and Caspase-3) were significantly suppressed. The combined therapy demonstrated a synergistic effect (combination index < 1). In conclusion, the results indicated that 10-HDA and ZnONPs have the potential to be a supplement or even an effective treatment to alleviate the adverse effects of lead poisoning. This is potentially attributed to their potent ameliorative actions against oxidation, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Samar R Saleh
- Bio-Screening and Preclinical Trial Lab, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Raheel G Agwah
- Bio-Screening and Preclinical Trial Lab, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22758, Egypt
| | - Ahmed Z Ghareeb
- Center of Excellence for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industry Development Center, City of Scientific Research & Technological Applications (SRTA-City), New Borg El Arab, Alexandria, Egypt
| | - Doaa A Ghareeb
- Bio-Screening and Preclinical Trial Lab, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
- Center of Excellence for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industry Development Center, City of Scientific Research & Technological Applications (SRTA-City), New Borg El Arab, Alexandria, Egypt.
- Research Projects Unit, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza Complex, Alexandria, 21648, Egypt.
| | - Adham M Maher
- Bio-Screening and Preclinical Trial Lab, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| |
Collapse
|
6
|
Sun F, Yu T, Zhang Y, Zhong X, Wang D, Li Y, Wang M, Zhang S, Yang T. AURKA inhibits the decidualization of the eutopic endometrium in endometriosis through nuclear factor-κB p65†. Biol Reprod 2025; 112:297-308. [PMID: 39673489 DOI: 10.1093/biolre/ioae176] [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: 09/09/2024] [Revised: 11/11/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024] Open
Abstract
Endometriosis is an estrogen dependent disease, which is related to infertility. Decidualization is a prerequisite for successful implantation of human embryos, and endometriosis affects the occurrence of decidualization. However, the mechanism that affects decidualization in endometriosis is not fully understood. Here, we find that Aurora kinase A (AURKA) is upregulated in the eutopic endometrium of endometriosis. AURKA inhibits the decidualization of stromal cells in the eutopic endometrium of endometriosis. Furthermore, in animal experiments, AURKA promotes endometriosis and inhibits decidualization in mice with endometriosis, leading to decreased expression of decidualization markers, such as prolactin, insulin-like growth factor-binding protein-1, and desmin. Afterwards, we find that nuclear factor-κB (NF-κB) p65 is a new substrate of AURKA. AURKA interacts with p65 to promote its phosphorylation and nuclear translocation. Meanwhile, AURKA enhances the protein stability of p65 by prolonging its half-life. In summary, AURKA inhibits the decidualization of the eutopic endometrium in patients with endometriosis by regulating p65, which may provide new ideas for improving decidualization defect in patients with endometriosis.
Collapse
Affiliation(s)
- Fangyuan Sun
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Ting Yu
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Ying Zhang
- Department of Gynecology, Zhucheng People's Hospital Affiliated to Shandong Second Medical University, Weifang, Shandong Province, People's Republic of China
| | - Xiaoyan Zhong
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Dan Wang
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Yuanyuan Li
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Mengxue Wang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Shucai Zhang
- Department of Emergency Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Tingting Yang
- Department of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| |
Collapse
|
7
|
Tian F, Sun S, Ge Z, Ge Y, Ge X, Shi Z, Qian X. Understanding the Anticancer Effects of Phytochemicals: From Molecular Docking to Anticarcinogenic Signaling. J Nutr 2025; 155:431-444. [PMID: 39581266 DOI: 10.1016/j.tjnut.2024.11.015] [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: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
As nontraditional nutrients, the biological activity of phytochemicals have been extensively studied for their antioxidant, anti-inflammatory, and apoptosis-promoting effects in various diseases. The general anticancer benefits of phytochemicals have been demonstrated in both basic researches and clinical trials. However, researchers understanding of how phytochemicals target cancer-related signaling pathways is still in its infancy. Molecular docking simulation analyses have yielded a large amount of cellular target molecules of phytochemicals. Herein, we review the potential signaling pathways that may be involved in the phytochemical-driven cancer benefits. We expect these findings to help in the design of potential cancer treatments designed by manipulating the binding modes and sites of these plant chemicals.
Collapse
Affiliation(s)
- Fuwei Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuhong Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zehe Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqian Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Alruhaimi RS, Hussein OE, Alnasser SM, Elbagory I, Alzoghaibi MA, Kamel EM, El Mohtadi M, Mahmoud AM. Haloxylon salicornicum Phytochemicals Suppress NF-κB, iNOS and Pro-Inflammatory Cytokines in Lipopolysaccharide-Induced Macrophages. Chem Biodivers 2025; 22:e202401623. [PMID: 39355861 DOI: 10.1002/cbdv.202401623] [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: 07/04/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/03/2024]
Abstract
Haloxylon salicornicum is traditionally used for the treatment of several disorders associated with inflammation. Despite it is a defense response against tissue injury and infections, inflammation can become a chronic condition that can negatively impact the body. This study investigated the effect of H. salicornicum phytochemicals nuclear factor-kappaB (NF-κB), inducible nitric oxide synthase (iNOS) and cytokines release by lipopolysaccharide (LPS)-challenged macrophages in vitro. The binding affinity of the tested phytochemical towards NF-κB and iNOS was investigated using molecular docking. Ten compounds (four coumarins, three sterols and three flavonoids) were isolated from the ethanolic extract of H. salicornicum. Treatment of LPS-challenged macrophages with the compounds resulted in remarkable decrease in NF-κB p65 and iNOS mRNA abundance. All compounds suppressed the production of nitric oxide (NO) and the pro-inflammatory cytokines (tumor necrosis factor (TNF)-α and interleukin (IL)-6) from macrophages challenged with LPS. Molecular docking revealed the ability of the isolated phytochemicals to bind NF-κB p65 and iNOS. In conclusion, H. salicornicum is a rich source of phytochemicals with anti-inflammatory properties. The anti-inflammatory efficacy of H. salicornicum phytoconstituents is mediated via their ability to modulate NF-κB and iNOS, and suppress the release of NO, TNF-α, and IL-6 from macrophages.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Omnia E Hussein
- Higher Technological Institute for Applied Health Sciences, Beni-Suef, Egypt
| | - Sulaiman M Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia
| | - Ibrahim Elbagory
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Mohammed A Alzoghaibi
- Physiology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Emadeldin M Kamel
- Organic Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | | | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK
| |
Collapse
|
9
|
da Silva A, Bobermin LD, Santos CL, de Souza Almeida RR, Lissner LJ, Dos Santos TM, Seady M, Leite MC, Wyse ATS, Gonçalves CA, Quincozes-Santos A. Glia-related Acute Effects of Risperidone and Haloperidol in Hippocampal Slices and Astrocyte Cultures from Adult Wistar Rats: A Focus on Inflammatory and Trophic Factor Release. Neurochem Res 2024; 50:22. [PMID: 39560678 DOI: 10.1007/s11064-024-04273-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 11/20/2024]
Abstract
Antipsychotics are drugs commonly prescribed to treat a variety of psychiatric conditions. They are classified as typical and atypical, depending on their affinity for dopaminergic and serotonergic receptors. Although neurons have been assumed to be the major mediators of the antipsychotic pharmacological effects, glia, particularly astrocytes, have emerged as important cellular targets for these drugs. In the present study, we investigated the effects of acute treatments with the antipsychotics risperidone and haloperidol of hippocampal slices and astrocyte cultures, focusing on neuron-glia communication and how antipsychotics act in astrocytes. For this, we obtained hippocampal slices and primary astrocyte cultures from 30-day-old Wistar rats and incubated them with risperidone or haloperidol (1 and 10 μM) for 30 min and 24 h, respectively. We evaluated metabolic and enzymatic activities, the glutathione level, the release of inflammatory and trophic factors, as well as the gene expression of signaling proteins. Haloperidol increased glucose metabolism; however, neither of the tested antipsychotics altered the glutathione content or glutamine synthetase and Na+K+-ATPase activities. Haloperidol induced a pro-inflammatory response and risperidone promoted an anti-inflammatory response, while both antipsychotics seemed to decrease trophic support. Haloperidol and risperidone increased Nrf2 and HO-1 gene expression, but only haloperidol upregulated NFκB and AMPK gene expression. Finally, astrocyte cultures confirmed the predominant effect of the tested antipsychotics on glia and their opposite effects on astrocytes. Therefore, antipsychotics cause functional alterations in the hippocampus. This information is important to drive future research for strategies to attenuate antipsychotics-induced neural dysfunction, focusing on glia.
Collapse
Affiliation(s)
- Amanda da Silva
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Camila Leite Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | | | - Tiago Marcon Dos Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Marina Seady
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Marina Concli Leite
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Carlos-Alberto Gonçalves
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências, Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação Em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
10
|
Shimizu Y, Yoshida T, Ito K, Terada K, Sasaki N, Honda E, Motomura K. Association Between Asian Flush and Satisfaction of Sleep via Alcohol Consumption Status in a Sample of Japanese Participants. Med Sci (Basel) 2024; 12:62. [PMID: 39584912 PMCID: PMC11587126 DOI: 10.3390/medsci12040062] [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: 09/02/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The reddening of the face and neck following alcohol consumption is known as Asian flush. Although genetic factors related to Asian flush have been reported to be inversely associated with duration of sleep, no study has reported an association between Asian flush and sleep satisfaction. METHODS A cross-sectional study of 3823 Japanese people, aged 20 to 64 years was conducted. The target population comprised general public resident monitors of Osaka Prefecture who were registered with an internet research company. RESULTS A significant inverse association was observed between Asian flush and satisfaction of sleep. The potential confounder-adjusted odds ratio (OR) and 95% confidence interval (CI) of satisfied sleep was 0.81 (0.69-0.96). The alcohol consumption status-specific analysis revealed essentially the same associations between consumers and non-consumers of alcohol. The adjusted ORs (95% CIs) were 0.81 (0.65-0.997) for non-consumers and 0.80 (0.61-1.05) for consumers of alcohol. CONCLUSION The genetic characteristics of physical reactions to alcohol exposure may influence sleep quality. One's alcohol consumption status may not influence the effects of having a lower tolerance to alcohol on sleep quality.
Collapse
Affiliation(s)
- Yuji Shimizu
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Tomokatsu Yoshida
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
| | - Keiko Ito
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
| | - Kumiko Terada
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
| | - Nagisa Sasaki
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
| | - Eiko Honda
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
| | - Kazushi Motomura
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan; (T.Y.); (K.I.); (K.T.); (N.S.); (E.H.); (K.M.)
- Department of Public Health, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Yan T, Sun J, Zhang Y, Wen C, Yang J. Enteromorpha prolifera Polysaccharide Alleviates Acute Alcoholic Liver Injury in C57 BL/6 Mice through the Gut-Liver Axis and NF-κB Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23258-23270. [PMID: 39404145 DOI: 10.1021/acs.jafc.4c05262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Enteromorpha prolifera polysaccharide (EP2) protection against acute alcoholic liver injury (AALI) in mice was investigated. By integration of physiological indicators, gut microbiota, and short-chain fatty acids (SCFAs), the mechanism of EP2 in alleviating AALI was disclosed. The results showed that EP2 significantly ameliorated alcohol-induced abnormal transaminase activities, liver and intestinal systemic inflammation, and intestinal environmental disorders. EP2 significantly reduces liver and serum LPS contents by 1.69-fold and 1.54-fold. Furthermore, inhibition of the NF-κB signaling pathway by EP2 reduced the production of proinflammatory cytokines such as TNF-α (1.83-fold), IL-6 (11.09-fold), and IL-1β (1.99-fold). EP2 restored SCFAs to normal levels by upregulating the abundance of beneficial bacteria (Colidextribacter, Ruminococcus, unclassified_Lachnospiraceae, and Akkermansia). The alleviation of AALI by EP2 occurs through protection of the intestinal mucosal barrier and reduction of LPS permeating in serum. The decrease in LPS inactivates the NF-κB signaling pathway and prevents inflammation. In short, EP2 regulates the gut-liver axis and inflammation, alleviating effects in AALI mice.
Collapse
Affiliation(s)
- Tingting Yan
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jinghe Sun
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Yuying Zhang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Chengrong Wen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jingfeng Yang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
12
|
Qi Y, Sun J, Wang H, Yu H, Jin X, Feng X, Wang Y. Effects of arsenic exposure on the PI3K/Akt/NF-κB signaling pathway in the hippocampus of offspring mice at different developmental stages. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116830. [PMID: 39111240 DOI: 10.1016/j.ecoenv.2024.116830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/02/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024]
Abstract
The primary purpose of present study was to explore the effects of arsenic exposure on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/nuclear transcription factor-κB (NF-κB) signaling pathway in the hippocampus of offspring mice at different developmental stages. Sodium arsenite (NaAsO2) at doses of 0, 15, 30 or 60 mg/L administered to female mice and their pups. The nuclear translocation levels of NF-κB were assessed by EMSA. Real-time RT-PCR was used to measure Akt, NF-κB and PI3K mRNA levels. Protein expressions of PI3K, p-Akt, inhibitor kappa B kinase (IKK), p-NF-κB, protein kinase A (PKA), inhibitor kappa B (IκB), and cAMP response element-binding protein (CREB) were measured by Western blot. Results disclosed that exposure to 60 mg/L NaAsO2 could suppress NF-κB levels of nuclear translocation of postnatal day (PND) 20 and PND 40 mice. Arsenic downregulated the transcriptional and translational levels of PI3K, Akt and NF-κB. Additionally, protein expressions of p-IKK, p-IκB, PKA and p-CREB also reduced. Taken together, results of present study indicated that arsenic could downregulate the PI3K/Akt/NF-κB signaling pathway, particularly on PND 40, which might be involved in the cognitive impairments.
Collapse
Affiliation(s)
- Yingying Qi
- Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China; Zhuhai Center for Chronic Disease Control(the Third Hospital of Zhuhai), People's Republic of China
| | - Jiaqi Sun
- Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China
| | - Huan Wang
- Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environment and Population Health of the Educational Department of Liaoning Province, Shenyang, People's Republic of China
| | - Haiyang Yu
- Key Laboratory of Environment and Population Health of the Educational Department of Liaoning Province, Shenyang, People's Republic of China; Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China
| | - Xiaoxia Jin
- Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environment and Population Health of the Educational Department of Liaoning Province, Shenyang, People's Republic of China
| | - Xu Feng
- Department of Health Statistics, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environment and Population Health of the Educational Department of Liaoning Province, Shenyang, People's Republic of China.
| |
Collapse
|
13
|
Shi Z, Zhang J, Ma H, Jing L. Network pharmacology and in vivo experimental studies reveal the protective effects of 6-hydroxygenistein against hypobaric hypoxia-induced brain injury. Heliyon 2024; 10:e36241. [PMID: 39253263 PMCID: PMC11382173 DOI: 10.1016/j.heliyon.2024.e36241] [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: 05/04/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Hypobaric hypoxia-induced brain injury (HHBI) is a progressive neurodegenerative disease that has still not been effectively treated. There are several different mechanisms involved in HHBI. Among them, oxidative stress and inflammation response predominate. 6-hydroxygenistein (4',5,6,7-tetrahydroxyisoflavone, 6-OHG) is a hydroxylated derivative of genistein with excellent antioxidant activity, however, the protective effects and underlying mechanisms against HHBI have not been clarified. In the present study, we aimed to explore the mechanisms of action of 6-OHG on HHBI using network pharmacology and experimental validation. Network pharmacology analysis revealed 186 candidate targets through the intersection of the targets of 6-OHG and related genes in HHBI, which were mainly enriched in oxidative stress and inflammation response. Moreover, key targets of 6-OHG against HHBI, namely Nrf2 and NF-κB, were screened and found to be closely related to oxidative stress and inflammation response. Subsequent in vivo experiments revealed that 6-OHG treatment attenuated oxidative stress and inflammation response, prevented energy disorder and apoptosis as well as maintained the BBB integrity in HHBI mice. In addition, 6-OHG administration up-regulated the expressions of Nrf2 and HO-1 and down-regulated the expressions of NF-κB and NLRP3, thereby inhibiting oxidative stress and inflammation response. Hence, the present study demonstrates that 6-OHG protects against HHBI by stimulating the Nrf2/HO-1 signaling pathway and suppressing the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Zhiqun Shi
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, China
| | - Jie Zhang
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Huiping Ma
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, China
| | - Linlin Jing
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Pharmacy, the 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, Gansu, 730050, China
| |
Collapse
|
14
|
Rosenbaum SR, Hughes CJ, Fields KM, Purdy SC, Gustafson A, Wolin A, Hampton D, Turner N, Ebmeier C, Costello JC, Ford HL. An EYA3/NF-κB/CCL2 signaling axis suppresses cytotoxic NK cells in the pre-metastatic niche to promote triple negative breast cancer metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606072. [PMID: 39211066 PMCID: PMC11360953 DOI: 10.1101/2024.07.31.606072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Patients with Triple Negative Breast Cancer (TNBC) exhibit high rates of metastases and poor prognoses. The Eyes absent (EYA) family of proteins are developmental transcriptional cofactors/phosphatases that are re-expressed and/or upregulated in numerous cancers. Herein, we demonstrate that EYA3 correlates with decreased survival in breast cancer, and that it strongly, and specifically, regulates metastasis via a novel mechanism that involves NF-kB signaling and an altered innate immune profile at the pre-metastatic niche (PMN). Remarkably, restoration of NF-kB signaling downstream of Eya3 knockdown (KD) restores metastasis without restoring primary tumor growth, isolating EYA3/NF-kB effects to the metastatic site. We show that secreted CCL2, regulated downstream of EYA3/NF-kB, specifically decreases cytotoxic NK cells in the PMN and that re-expression of Ccl2 in Eya3 -KD cells is sufficient to rescue activation/levels of cytotoxic NK cells in vitro and at the PMN, where EYA3-mediated decreases in cytotoxic NK cells are required for metastatic outgrowth. Importantly, analysis of public breast cancer datasets uncovers a significant correlation of EYA3 with NF-kB/CCL2, underscoring the relevance of EYA3/NF-kB/CCL2 to human disease. Our findings suggest that inhibition of EYA3 could be a powerful means to re-activate the innate immune response at the PMN, inhibiting TNBC metastasis. Significance EYA3 promotes metastasis of TNBC cells by promoting NF-kB-mediated CCL2 expression and inhibiting cytotoxic NK cells at the pre-metastatic niche, highlighting a potential therapeutic target in this subset of breast cancer.
Collapse
|
15
|
Yang F, Dai Z, Xue MY, Chen XY, Liu J, Wang L, Xu LL, Di B. Identification and Validation of PKR as a Direct Target for the Novel Sulfonamide-Substituted Tetrahydroquinoline Nonselective Inhibitor of the NLRP3 Inflammasome. J Med Chem 2024; 67:10168-10189. [PMID: 38855903 DOI: 10.1021/acs.jmedchem.4c00343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The NLRP3 inflammasome is a critical component of the innate immune system. The persistent abnormal activation of the NLRP3 inflammasome is implicated in numerous human diseases. Herein, sulfonamide-substituted tetrahydroquinoline derivative S-9 was identified as the most promising NLRP3 inhibitor, without obvious cytotoxicity. In vitro, S-9 inhibited the priming and activation stages of the NLRP3 inflammasome. Incidentally, we also observed that S-9 had inhibitory effects on the NLRC4 and AIM2 inflammasomes. To elucidate the multiple anti-inflammatory activities of S-9, photoaffinity probe P-2, which contained a photoaffinity label and a functional handle, was developed for target identification by chemical proteomics. We identified PKR as a novel target of S-9 in addition to NLRP3 by target fishing. Furthermore, S-9 exhibited a significant anti-neuroinflammatory effect in vivo. In summary, our findings show that S-9 is a promising lead compound targeting both PKR and NLRP3 that could emerge as a molecular tool for treating inflammasome-related diseases.
Collapse
Affiliation(s)
- Fan Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Zhen Dai
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Ming-Yue Xue
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Yi Chen
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Juan Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Li Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Bin Di
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
16
|
Li S, Sun J, Zhang BW, Yang L, Wan YC, Chen BB, Xu N, Xu QR, Fan J, Shang JN, Li R, Yu CG, Xi Y, Chen S. ATG5 attenuates inflammatory signaling in mouse embryonic stem cells to control differentiation. Dev Cell 2024; 59:882-897.e6. [PMID: 38387460 DOI: 10.1016/j.devcel.2024.01.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/13/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Abstract
Attenuated inflammatory response is a property of embryonic stem cells (ESCs). However, the underlying mechanisms are unclear. Moreover, whether the attenuated inflammatory status is involved in ESC differentiation is also unknown. Here, we found that autophagy-related protein ATG5 is essential for both attenuated inflammatory response and differentiation of mouse ESCs and that attenuation of inflammatory signaling is required for mouse ESC differentiation. Mechanistically, ATG5 recruits FBXW7 to promote ubiquitination and proteasome-mediated degradation of β-TrCP1, resulting in the inhibition of nuclear factor κB (NF-κB) signaling and inflammatory response. Moreover, differentiation defects observed in ATG5-depleted mouse ESCs are due to β-TrCP1 accumulation and hyperactivation of NF-κB signaling, as loss of β-TrCP1 and inhibition of NF-κB signaling rescued the differentiation defects. Therefore, this study reveals a previously uncharacterized mechanism maintaining the attenuated inflammatory response in mouse ESCs and further expands the understanding of the biological roles of ATG5.
Collapse
Affiliation(s)
- Sheng Li
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China; School of Forensic Sciences and Laboratory Medicine, Jining Medical University, Jining 272067, Shandong, China
| | - Jin Sun
- School of Laboratory Animal & Shandong Laboratory Animal Center, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Bo-Wen Zhang
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Lu Yang
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Ying-Cui Wan
- School of Laboratory Animal & Shandong Laboratory Animal Center, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Bei-Bei Chen
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Nan Xu
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Qian-Ru Xu
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Juan Fan
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Jia-Ni Shang
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Rui Li
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Chen-Ge Yu
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China
| | - Yan Xi
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China; Zhongzhou Laboratory, Kaifeng 475004, Henan, China.
| | - Su Chen
- Laboratory of Molecular and Cellular Biology, Institute of Metabolism and Health, School of Basic Medical Sciences, Department of General Surgery of Huaihe Hospital, Henan University, Kaifeng 475004, Henan, China; Zhongzhou Laboratory, Kaifeng 475004, Henan, China.
| |
Collapse
|
17
|
Chen S, Lin T, Basu R, Ritchey J, Wang S, Luo Y, Li X, Pei D, Kara LB, Cheng X. Design of target specific peptide inhibitors using generative deep learning and molecular dynamics simulations. Nat Commun 2024; 15:1611. [PMID: 38383543 PMCID: PMC10882002 DOI: 10.1038/s41467-024-45766-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/04/2024] [Indexed: 02/23/2024] Open
Abstract
We introduce a computational approach for the design of target-specific peptides. Our method integrates a Gated Recurrent Unit-based Variational Autoencoder with Rosetta FlexPepDock for peptide sequence generation and binding affinity assessment. Subsequently, molecular dynamics simulations are employed to narrow down the selection of peptides for experimental assays. We apply this computational strategy to design peptide inhibitors that specifically target β-catenin and NF-κB essential modulator. Among the twelve β-catenin inhibitors, six exhibit improved binding affinity compared to the parent peptide. Notably, the best C-terminal peptide binds β-catenin with an IC50 of 0.010 ± 0.06 μM, which is 15-fold better than the parent peptide. For NF-κB essential modulator, two of the four tested peptides display substantially enhanced binding compared to the parent peptide. Collectively, this study underscores the successful integration of deep learning and structure-based modeling and simulation for target specific peptide design.
Collapse
Affiliation(s)
- Sijie Chen
- College of Pharmacy, The Ohio State University, 281 W Lane Ave, Columbus, OH, USA
| | - Tong Lin
- Mechanical Engineering Department, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA, USA
- Machine Learning Department, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA, USA
| | - Ruchira Basu
- Department of Chemistry and Biochemistry, The Ohio State University, 281 W Lane Ave, Columbus, OH, USA
| | - Jeremy Ritchey
- Department of Chemistry and Biochemistry, The Ohio State University, 281 W Lane Ave, Columbus, OH, USA
| | - Shen Wang
- College of Pharmacy, The Ohio State University, 281 W Lane Ave, Columbus, OH, USA
| | - Yichuan Luo
- Electrical and Computer Engineering Department, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA, USA
| | - Xingcan Li
- Department of Radiology, Affiliated Hospital and Medical School of Nantong University, 20 West Temple Road, Nantong, Jiangsu, China
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 281 W Lane Ave, Columbus, OH, USA.
| | - Levent Burak Kara
- Mechanical Engineering Department, Carnegie Mellon University, 5000 Forbes Ave, Pittsburgh, PA, USA.
| | - Xiaolin Cheng
- College of Pharmacy, The Ohio State University, 281 W Lane Ave, Columbus, OH, USA.
- Translational Data Analytics Institute, The Ohio State University, 1760 Neil Ave, Columbus, OH, USA.
| |
Collapse
|
18
|
Gayatri V, Krishna Prasad M, Mohandas S, Nagarajan S, Kumaran K, Ramkumar KM. Crosstalk between inflammasomes, inflammation, and Nrf2: Implications for gestational diabetes mellitus pathogenesis and therapeutics. Eur J Pharmacol 2024; 963:176241. [PMID: 38043778 DOI: 10.1016/j.ejphar.2023.176241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
The role of inflammasomes in gestational diabetes mellitus (GDM) has emerged as a critical area of research in recent years. Inflammasomes, key components of the innate immune system, are now recognized for their involvement in the pathogenesis of GDM. Activation of inflammasomes in response to various triggers during pregnancy can produce pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and interleukin-18 (IL-18), contributing to systemic inflammation and insulin resistance. This dysregulation not only impacts maternal health but also poses significant risks to fetal development and long-term health outcomes. Understanding the intricate interplay between inflammasomes and GDM holds promise for developing novel therapeutic strategies and interventions to mitigate the adverse effects of this condition on both mothers and their offspring. Researchers have elucidated that targeting inflammasomes using anti-inflammatory drugs and compounds can effectively reduce inflammation in GDM. Furthermore, the addition of nuclear factor erythroid 2-related factor 2 (Nrf2) to this complex mechanism opens novel avenues for therapeutics. The antioxidant properties of Nrf2 may potentially suppress inflammasome activation in GDM. This comprehensive review investigates the intricate relationship between inflammasomes and GDM, emphasizing the pivotal role of inflammation in its pathogenesis. It also sheds light on potential therapeutic strategies targeting inflammasome activation and explores the role of Nrf2 in mitigating inflammation in GDM.
Collapse
Affiliation(s)
- Vijaya Gayatri
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Murali Krishna Prasad
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Sundhar Mohandas
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Sanjushree Nagarajan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kriya Kumaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
19
|
Albekairi TH, Alanazi MM, Ansari MA, Nadeem A, Attia SM, Bakheet SA, Al-Mazroua HA, Aldossari AA, Almanaa TN, Alwetaid MY, Alqinyah M, Alnefaie HO, Ahmad SF. Cadmium exposure exacerbates immunological abnormalities in a BTBR T + Itpr3 tf/J autistic mouse model by upregulating inflammatory mediators in CD45R-expressing cells. J Neuroimmunol 2024; 386:578253. [PMID: 38064869 DOI: 10.1016/j.jneuroim.2023.578253] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental illness characterized by behavior, learning, communication, and social interaction abnormalities in various situations. Individuals with impairments usually exhibit restricted and repetitive actions. The actual cause of ASD is yet unknown. It is believed, however, that a mix of genetic and environmental factors may play a role in its development. Certain metals have been linked to the development of neurological diseases, and the prevalence of ASD has shown a positive association with industrialization. Cadmium chloride (Cd) is a neurotoxic chemical linked to cognitive impairment, tremors, and neurodegenerative diseases. The BTBR T+ Itpr3tf/J (BTBR) inbred mice are generally used as a model for ASD and display a range of autistic phenotypes. We looked at how Cd exposure affected the signaling of inflammatory mediators in CD45R-expressing cells in the BTBR mouse model of ASD. In this study, we looked at how Cd affected the expression of numerous markers in the spleen, including IFN-γ, IL-6, NF-κB p65, GM-CSF, iNOS, MCP-1, and Notch1. Furthermore, we investigated the effect of Cd exposure on the expression levels of numerous mRNA molecules in brain tissue, including IFN-γ, IL-6, NF-κB p65, GM-CSF, iNOS, MCP-1, and Notch1. The RT-PCR technique was used for this analysis. Cd exposure increased the number of CD45R+IFN-γ+, CD45R+IL-6+, CD45R+NF-κB p65+, CD45R+GM-CSF+, CD45R+GM-CSF+, CD45R+iNOS+, and CD45R+Notch1+ cells in the spleen of BTBR mice. Cd treatment also enhanced mRNA expression in brain tissue for IFN-γ, IL-6, NF-κB, GM-CSF, iNOS, MCP-1, and Notch1. In general, Cd increases the signaling of inflammatory mediators in BTBR mice. This study is the first to show that Cd exposure causes immune function dysregulation in the BTBR ASD mouse model. As a result, our study supports the role of Cd exposure in the development of ASD.
Collapse
Affiliation(s)
- Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah A Aldossari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Y Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hajar O Alnefaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
20
|
Joung EJ, Lee MK, Lee M, Gwon M, Shin T, Ryu H, Jeong HH, Kim MJ, Van JY, Kim JI, Choi J, Jung WK, Kim HR, Lee B. Sargachromenol Attenuates Inflammatory Responses by Regulating NF-κB and Nrf2 Pathways in RAW 264.7 Cells and LPS-treated Mice. PLANTA MEDICA 2024; 90:25-37. [PMID: 37848042 DOI: 10.1055/a-2180-1338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
This study aims to explore the anti-inflammatory mechanisms of sargachromenol in both RAW 264.7 cells and lipopolysaccharide (LPS)-treated mice, as previous reports have suggested that sargachromenol possesses anti-aging, anti-inflammatory, antioxidant, and neuroprotective properties. Although the precise mechanism behind its anti-inflammatory activity remains unclear, pretreatment with sargachromenol effectively reduced the production of nitric oxide, prostaglandin E2, and interleukin (IL)-1β in LPS-stimulated RAW 264.7 cells by inhibiting cyclooxygenase-2. Moreover, sargachromenol inhibited the activation of nuclear factor-κB (NF-κB) by preventing the degradation of the inhibitor of κB-α (IκB-α) and inhibiting protein kinase B (Akt) phosphorylation in LPS-stimulated cells. We also found that sargachromenol induced the production of heme oxygenase-1 (HO-1) by activating the nuclear transcription factor erythroid-2-related factor 2 (Nrf2). In LPS-treated mice, oral administration of sargachromenol effectively reduced the levels of IL-1β, IL-6, and tumor necrosis factor-α (TNF-α) in the serum, suggesting its ability to suppress the production of inflammatory mediators by inhibiting the Akt/NF-κB pathway and upregulating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Eun-Ji Joung
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Min-Kyeong Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Minsup Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Louisiana, United States
| | - Misung Gwon
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Taisun Shin
- Division of Food and Nutrition, Chonnam National University, Gwangju, Republic of Korea
| | - Heeyeon Ryu
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Hyeon Hak Jeong
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| | - Myeong-Jin Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Ji Yun Van
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| | - Jae-Il Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Jinkyung Choi
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Won-Kyo Jung
- Division of Biomedical Engineering and Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Hyeung-Rak Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Republic of Korea
- Department of Smart Green Technology Engineering, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
21
|
Sherekar S, Todankar CS, Viswanathan GA. Modulating the dynamics of NFκB and PI3K enhances the ensemble-level TNFR1 signaling mediated apoptotic response. NPJ Syst Biol Appl 2023; 9:57. [PMID: 37973854 PMCID: PMC10654705 DOI: 10.1038/s41540-023-00318-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
Cell-to-cell variability during TNFα stimulated Tumor Necrosis Factor Receptor 1 (TNFR1) signaling can lead to single-cell level pro-survival and apoptotic responses. This variability stems from the heterogeneity in signal flow through intracellular signaling entities that regulate the balance between these two phenotypes. Using systematic Boolean dynamic modeling of a TNFR1 signaling network, we demonstrate that the signal flow path variability can be modulated to enable cells favour apoptosis. We developed a computationally efficient approach "Boolean Modeling based Prediction of Steady-state probability of Phenotype Reachability (BM-ProSPR)" to accurately predict the network's ability to settle into different phenotypes. Model analysis juxtaposed with the experimental observations revealed that NFκB and PI3K transient responses guide the XIAP behaviour to coordinate the crucial dynamic cross-talk between the pro-survival and apoptotic arms at the single-cell level. Model predicted the experimental observations that ~31% apoptosis increase can be achieved by arresting Comp1 - IKK* activity which regulates the NFκB and PI3K dynamics. Arresting Comp1 - IKK* activity causes signal flow path re-wiring towards apoptosis without significantly compromising NFκB levels, which govern adequate cell survival. Priming an ensemble of cancerous cells with inhibitors targeting the specific interaction involving Comp1 and IKK* prior to TNFα exposure could enable driving them towards apoptosis.
Collapse
Affiliation(s)
- Shubhank Sherekar
- Department of Chemical Engineering, Indian Institute of Technology Bombay Powai, Mumbai, 400076, India
| | - Chaitra S Todankar
- Department of Chemical Engineering, Indian Institute of Technology Bombay Powai, Mumbai, 400076, India
| | - Ganesh A Viswanathan
- Department of Chemical Engineering, Indian Institute of Technology Bombay Powai, Mumbai, 400076, India.
| |
Collapse
|
22
|
Liu S, Zhang F, Liang Y, Wu G, Liu R, Li X, Saw PE, Yang Z. Nanoparticle (NP)-mediated APOC1 silencing to inhibit MAPK/ERK and NF-κB pathway and suppress breast cancer growth and metastasis. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2451-2465. [PMID: 37668862 DOI: 10.1007/s11427-022-2329-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/14/2023] [Indexed: 09/06/2023]
Abstract
Breast cancer is one of the most common malignant tumors with high mortality and poor prognosis in women. There is an urgent need to discover new therapeutic targets for breast cancer metastasis. Herein, we identified that Apolipoprotein C1 (APOC1) was up-regulated in primary tumor of breast cancer patient that recurrence and metastasis by immunohistochemistry (IHC). Kaplan-Meier Plotter database showed that high levels of APOC1 in breast cancer patients were strongly associated with worse overall survival (OS) and relapse-free survival (RFS). Mechanistically, APOC1 silencing significantly inhibits MAPK/ERK kinase pathway and restrains the NF-κB to decrease the transcription of target genes related to growth and metastasis in vitro. Based on this regulatory mechanism, we developed these findings into potential therapeutic drugs, glutathione (GSH) responsive nano-particles (NPs) were used for systemic APOC1 siRNA delivery, NPs (siAPOC1) silenced APOC1 expression, and subsequently resulted in positive anti-tumor effects in orthotopic and liver metastasis models in vivo. Taken together, GSH responsive NP-mediated siAPOC1 delivery was proved to be effective in regulating growth and metastasis in multiple tumor models. These findings show that APOC1 could be a potential biomarker to predict the prognosis of breast cancer patients and NP-mediated APOC1 silencing could be new strategies for exploration of new treatments for breast cancer metastasis.
Collapse
Affiliation(s)
- Shaomin Liu
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China
| | - Fengqian Zhang
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Yixia Liang
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Guo Wu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, 421001, China
| | - Rong Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, 421001, China
| | - Xiuling Li
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Phei Er Saw
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| | - Zhonghan Yang
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
23
|
Datsyuk JK, Paudel KR, Rajput R, Kokkinis S, El Sherkawi T, Singh SK, Gupta G, Chellappan DK, Yeung S, Hansbro PM, Oliver BGG, Santos HA, Dua K, De Rubis G. Emerging applications and prospects of NFκB decoy oligodeoxynucleotides in managing respiratory diseases. Chem Biol Interact 2023; 385:110737. [PMID: 37774998 DOI: 10.1016/j.cbi.2023.110737] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Chronic respiratory diseases like asthma and Chronic Obstructive Pulmonary Disease (COPD) have been a burden to society for an extended period. Currently, there are only preventative treatments in the form of mono- or multiple-drug therapy available to patients who need to utilize it daily. Hence, throughout the years there has been a substantial amount of research in understanding what causes inflammation in the context of these diseases. For example, the transcription factor NFκB has a pivotal role in causing chronic inflammation. Subsequent research has been exploring ways to block the activation of NFκB as a potential therapeutic strategy for many inflammatory diseases. One of the possible ways through which this is probable is the utilisation of decoy oligodeoxynucleotides, which are synthetic, short, single-stranded DNA fragments that mimic the consensus binding site of a targeted transcription factor, thereby functionally inactivating it. However, limitations to the implementation of decoy oligodeoxynucleotides include their rapid degradation by intracellular nucleases and the lack of targeted tissue specificity. An advantageous approach to overcome these limitations involves using nanoparticles as a vessel for drug delivery. In this review, all of those key elements will be explored as to how they come together as an application to treat chronic inflammation in respiratory diseases.
Collapse
Affiliation(s)
- Jessica Katrine Datsyuk
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Rashi Rajput
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Sofia Kokkinis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Tammam El Sherkawi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Jaipur, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000, Malaysia
| | - Stewart Yeung
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Philip Michael Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia
| | - Brian Gregory George Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, New South Wales, Australia; School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Hélder A Santos
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
24
|
Vujovic F, Shepherd CE, Witting PK, Hunter N, Farahani RM. Redox-Mediated Rewiring of Signalling Pathways: The Role of a Cellular Clock in Brain Health and Disease. Antioxidants (Basel) 2023; 12:1873. [PMID: 37891951 PMCID: PMC10604469 DOI: 10.3390/antiox12101873] [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: 09/11/2023] [Revised: 10/14/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Metazoan signalling pathways can be rewired to dampen or amplify the rate of events, such as those that occur in development and aging. Given that a linear network topology restricts the capacity to rewire signalling pathways, such scalability of the pace of biological events suggests the existence of programmable non-linear elements in the underlying signalling pathways. Here, we review the network topology of key signalling pathways with a focus on redox-sensitive proteins, including PTEN and Ras GTPase, that reshape the connectivity profile of signalling pathways in response to an altered redox state. While this network-level impact of redox is achieved by the modulation of individual redox-sensitive proteins, it is the population by these proteins of critical nodes in a network topology of signal transduction pathways that amplifies the impact of redox-mediated reprogramming. We propose that redox-mediated rewiring is essential to regulate the rate of transmission of biological signals, giving rise to a programmable cellular clock that orchestrates the pace of biological phenomena such as development and aging. We further review the evidence that an aberrant redox-mediated modulation of output of the cellular clock contributes to the emergence of pathological conditions affecting the human brain.
Collapse
Affiliation(s)
- Filip Vujovic
- IDR/Westmead Institute for Medical Research, Sydney, NSW 2145, Australia; (F.V.); (N.H.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Paul K. Witting
- Redox Biology Group, Charles Perkins Centre, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Neil Hunter
- IDR/Westmead Institute for Medical Research, Sydney, NSW 2145, Australia; (F.V.); (N.H.)
| | - Ramin M. Farahani
- IDR/Westmead Institute for Medical Research, Sydney, NSW 2145, Australia; (F.V.); (N.H.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
25
|
Plantone D, Primiano G, Righi D, Romano A, Luigetti M, De Stefano N. Current Evidence Supporting the Role of Immune Response in ATTRv Amyloidosis. Cells 2023; 12:2383. [PMID: 37830598 PMCID: PMC10572348 DOI: 10.3390/cells12192383] [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: 08/16/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Hereditary transthyretin (ATTRv) amyloidosis with polyneuropathy, also known as familial amyloid polyneuropathy (FAP), represents a progressive, heterogeneous, severe, and multisystemic disease caused by pathogenic variants in the TTR gene. This autosomal-dominant neurogenetic disorder has an adult onset with variable penetrance and an inconstant phenotype, even among subjects carrying the same mutation. Historically, ATTRv amyloidosis has been viewed as a non-inflammatory disease, mainly due to the absence of any mononuclear cell infiltration in ex vivo tissues; nevertheless, a role of inflammation in its pathogenesis has been recently highlighted. The immune response may be involved in the development and progression of the disease. Fibrillary TTR species bind to the receptor for advanced glycation end products (RAGE), probably activating the nuclear factor κB (NF-κB) pathway. Moreover, peripheral blood levels of several cytokines, including interferon (IFN)-gamma, IFN-alpha, IL-6, IL-7, and IL-33, are altered in the course of the disease. This review summarizes the current evidence supporting the role of the immune response in ATTRv amyloidosis, from the pathological mechanisms to the possible therapeutic implications.
Collapse
Affiliation(s)
- Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; (D.R.); (N.D.S.)
| | - Guido Primiano
- Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.R.); (M.L.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Delia Righi
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; (D.R.); (N.D.S.)
| | - Angela Romano
- Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.R.); (M.L.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Luigetti
- Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.R.); (M.L.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; (D.R.); (N.D.S.)
| |
Collapse
|
26
|
Jiang L, Liu T, Lyu K, Chen Y, Lu J, Wang X, Long L, Li S. Inflammation-related signaling pathways in tendinopathy. Open Life Sci 2023; 18:20220729. [PMID: 37744452 PMCID: PMC10512452 DOI: 10.1515/biol-2022-0729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
Tendon is a connective tissue that produces movement by transmitting the force produced by muscle contraction to the bones. Most tendinopathy is caused by prolonged overloading of the tendon, leading to degenerative disease of the tendon. When overloaded, the oxygen demand of tenocytes increases, and the tendon structure is special and lacks blood supply, which makes it easier to form an oxygen-deficient environment in tenocytes. The production of reactive oxygen species due to hypoxia causes elevation of inflammatory markers in the tendon, including PGE2, IL-1β, and TNF-α. In the process of tendon healing, inflammation is also a necessary stage. The inflammatory environment formed by cytokines and various immune cells play an important role in the clearance of necrotic material, the proliferation of tenocytes, and the production of collagen fibers. However, excessive inflammation can lead to tendon adhesions and hinder tendon healing. Some important and diverse biological functions of the body originate from intercellular signal transduction, among which cytokine mediation is an important way of signal transduction. In particular, NF-κB, NLRP3, p38/MAPK, and signal transducer and activator of transcription 3, four common signaling pathways in tendinopathy inflammatory response, play a crucial role in the regulation and transcription of inflammatory factors. Therefore, summarizing the specific mechanisms of inflammatory signaling pathways in tendinopathy is of great significance for an in-depth understanding of the inflammatory response process and exploring how to inhibit the harmful part of the inflammatory response and promote the beneficial part to improve the healing effect of the tendon.
Collapse
Affiliation(s)
- Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Tianzhu Liu
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoqiang Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Longhai Long
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, 210000, China
| |
Collapse
|
27
|
Anoushirvani AA, Jafarian Yazdi A, Amirabadi S, Asouri SA, Shafabakhsh R, Sheida A, Hosseini Khabr MS, Jafari A, Tamehri Zadeh SS, Hamblin MR, Kalantari L, Talaei Zavareh SA, Mirzaei H. Role of non-coding RNAs in neuroblastoma. Cancer Gene Ther 2023; 30:1190-1208. [PMID: 37217790 DOI: 10.1038/s41417-023-00623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/25/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
Neuroblastoma is known as the most prevalent extracranial malignancy in childhood with a neural crest origin. It has been widely accepted that non-coding RNAs (ncRNAs) play important roles in many types of cancer, including glioma and gastrointestinal cancers. They may regulate the cancer gene network. According to recent sequencing and profiling studies, ncRNAs genes are deregulated in human cancers via deletion, amplification, abnormal epigenetic, or transcriptional regulation. Disturbances in the expression of ncRNAs may act either as oncogenes or as anti-tumor suppressor genes, and can lead to the induction of cancer hallmarks. ncRNAs can be secreted from tumor cells inside exosomes, where they can be transferred to other cells to affect their function. However, these topics still need more study to clarify their exact roles, so the present review addresses different roles and functions of ncRNAs in neuroblastoma.
Collapse
Affiliation(s)
- Ali Arash Anoushirvani
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sanaz Amirabadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX: 15179/64311, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran.
| |
Collapse
|
28
|
Chen M, He X, Sun Z, Huo X, Hou Y, Xu X, Wu H, Shi L, Ma G. Natural carrier-free self-assembled diterpene nanoparticles with its efficient anti-inflammation through the inhibition of NF-κB pathway for accelerated wound healing. Biomed Pharmacother 2023; 165:115041. [PMID: 37356374 DOI: 10.1016/j.biopha.2023.115041] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023] Open
Abstract
Nanoscience has set off a wave in biomedicine to improve the performance of drugs in recent years, but additional materials are usually required for supramolecular nanoconstruction, undoubtedly increasing the health risks. Herein, we discovered a novel diterpene supramolecular self-assembly system without additional chemicals, Nepebracteatalic Acid nanoparticles (NA NPs), mediated through hydrogen bond, hydrophobic and electrostatic interaction. NA NPs performed sustained release behavior, lower expression levels for IL-6 and TNF-α than clinical anti-inflammatory drug Indometacin. Furthermore, the effect of NA NPs on the related protein p65 expression levels of nuclear factor-κB (NFκB) signaling pathway is quantified to confirm the enhanced anti-inflammatory property based on the self-assembly strategy. Meanwhile, the prepared nanoparticles have good biocompatibility which ensures outstanding inflammation inhibition, collagen deposition, angiogenesis during wound healing. This work opens up new prospects that carrier-free nanoparticles from NPMs have great potential to exert clinical application value, meanwhile providing reference for developing green nanoscience.
Collapse
Affiliation(s)
- Meiying Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xiaoli He
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Zhaocui Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xiaowei Huo
- College of Pharmaceutical Sciences, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, No. 180, East Wusi Road, Baoding 071002, China
| | - Yong Hou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xudong Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Haifeng Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Leiling Shi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; Xinjiang Institute of Chinese and Ethnic Medicine, No. 88, Weisheng Lane, Xinmin Road, Tianshan District, Urumqi 830002, China.
| | - Guoxu Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China.
| |
Collapse
|
29
|
Meng F, Zhou Y, Wagner A, Bülow JM, Köhler K, Neunaber C, Bundkirchen K, Relja B. Impact of age on liver damage, inflammation, and molecular signaling pathways in response to femoral fracture and hemorrhage. Front Immunol 2023; 14:1239145. [PMID: 37691959 PMCID: PMC10484338 DOI: 10.3389/fimmu.2023.1239145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Background Trauma causes disability and mortality globally, leading to fractures and hemorrhagic shock. This can trigger an irregular inflammatory response that damages remote organs, including liver. Aging increases the susceptibility to dysregulated immune responses following trauma, raising the risk of organ damage, infections, and higher morbidity and mortality in elderly patients. This study investigates how aging affects liver inflammation and damage post-trauma. Methods 24 male C57BL/6J mice were randomly divided into four groups. Twelve young (17-26 weeks) and 12 aged (64-72 weeks) mice were included. Mice further underwent either hemorrhagic shock (trauma/hemorrhage, TH), and femoral fracture (osteotomy) with external fixation (Fx) (THFx, n=6) or sham procedures (n=6). After 24 hours, mice were sacrificed. Liver injury and apoptosis were evaluated using hematoxylin-eosin staining and activated caspase-3 immunostaining. CXCL1 and infiltrating polymorphonuclear leukocytes (PMNL) in the liver were assessed by immunostaining, and concentrations of CXCL1, TNF, IL-1β, and IL-10 in the liver tissue were determined by ELISA. Gene expression of Tnf, Cxcl1, Il-1β, and Cxcl2 in the liver tissue was determined by qRT-PCR. Finally, western blot was used to determine protein expression levels of IκBα, Akt, and their phosphorylated forms. Results THFx caused liver damage and increased presence of active caspase-3-positive cells compared to the corresponding sham group. THFx aged group had more severe liver injury than the young group. CXCL1 and PMNL levels were significantly higher in both aged groups, and THFx caused a greater increase in CXCL and PMNL levels in aged compared to the young group. Pro-inflammatory TNF and IL-1β levels were elevated in aged groups, further intensified by THFx. Anti-inflammatory IL-10 levels were lower in aged groups. Tnf and Cxcl1 gene expression was enhanced in the aged sham group. Phosphorylation ratio of IκBα was significantly increased in the aged sham group versus young sham group. THFx-induced IκBα phosphorylation in the young group was significantly reduced in the aged THFx group. Akt phosphorylation was significantly reduced in the THFx aged group compared to the THFx young group. Conclusion The findings indicate that aging may lead to increased vulnerability to liver injury and inflammation following trauma due to dysregulated immune responses.
Collapse
Affiliation(s)
- Fanshuai Meng
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
- Department of Trauma and Reconstructive Surgery, Uniklinik RWTH Aachen, Aachen, Germany
| | - Yuzhuo Zhou
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Alessa Wagner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Maria Bülow
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Claudia Neunaber
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Katrin Bundkirchen
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Borna Relja
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
30
|
Dai Y, Ma S, Zhu Y, Gontcharov AA, Liu Y, Wang Q. Immunomodulatory Effect of Flammulina rossica Fermentation Extract on Healthy and Immunosuppressed Mice. Molecules 2023; 28:5825. [PMID: 37570797 PMCID: PMC10421243 DOI: 10.3390/molecules28155825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Flammulina rossica fermentation extract (FREP) was obtained by ethanol precipitation of the fermentation broth. The molecular weight of FREP is 28.52 kDa, and it mainly contains active ingredients such as polysaccharides, proteins, reducing sugars, and 16 amino acids. Among them, the polysaccharides were mannose, glucose, galactose, arabinose, and fucose and possessed β-glycosidic bonds. Furthermore, the immunoregulatory activities of FREP were investigated in vivo. The results demonstrated that FREP could increase the counts of CD4+ T lymphocytes and the ratio of CD4+/CD8+ in a dose-dependent manner in healthy mice. In addition, FREP significantly increased serum cytokines, including IL-2, IL-8, IL-10, IL-12, IL-6, IL-1β, INF-γ, C-rection protein, and TNF-α, and promoted splenocyte proliferation in healthy mice. Finally, FREP could restore the counts of white blood cells, red blood cells, secretory immunoglobulin A, and antibody-forming cells and significantly promote the serum haemolysin level in mice treated with cyclophosphamide. The findings indicated that FREP possessed immunoregulatory activity in healthy mice and could improve the immune functions in immunosuppressive mice. Therefore, FREP could be exploited as an immunomodulatory agent and potential immunotherapeutic medicine for patients with inadequate immune function.
Collapse
Affiliation(s)
- Yingdi Dai
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (S.M.); (Y.Z.)
- College of Plant Protection, Jilin Agricultural University, Changchun 130012, China
| | - Sijia Ma
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (S.M.); (Y.Z.)
- College of Plant Protection, Jilin Agricultural University, Changchun 130012, China
| | - Yanyan Zhu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (S.M.); (Y.Z.)
| | - Andrey A. Gontcharov
- Institute of Biology and Soil Science, FEB RAS, 100-Letia Vladivostoka Prospect, 159, Vladivostok 690022, Russia;
| | - Yang Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (S.M.); (Y.Z.)
- College of Plant Protection, Jilin Agricultural University, Changchun 130012, China
| | - Qi Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (S.M.); (Y.Z.)
| |
Collapse
|
31
|
Jiang Y, Zhang J, Shi C, Li X, Jiang Y, Mao R. NF- κB: a mediator that promotes or inhibits angiogenesis in human diseases? Expert Rev Mol Med 2023; 25:e25. [PMID: 37503730 DOI: 10.1017/erm.2023.20] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) signaling pathway, which is conserved in invertebrates, plays a significant role in human diseases such as inflammation-related diseases and carcinogenesis. Angiogenesis refers to the growth of new capillary vessels derived from already existing capillaries and postcapillary venules. Maintaining normal angiogenesis and effective vascular function is a prerequisite for the stability of organ tissue function, and abnormal angiogenesis often leads to a variety of diseases. It has been suggested that NK-κB signalling molecules under pathological conditions play an important role in vascular differentiation, proliferation, apoptosis and tumourigenesis by regulating the transcription of multiple target genes. Many NF-κB inhibitors are being tested in clinical trials for cancer treatment and their effect on angiogenesis is summarised. In this review, we will summarise the role of NF-κB signalling in various neovascular diseases, especially in tumours, and explore whether NF-κB can be used as an attack target or activation medium to inhibit tumour angiogenesis.
Collapse
Affiliation(s)
- Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, 30Tongyang North Road, Pingchao Town, Nantong 226361, Jiangsu, People's Republic of China
| | - Conglin Shi
- Department of Pathogenic Biology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Xingjuan Li
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Yongying Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| |
Collapse
|
32
|
Shimizu Y. Progression of Carotid Intima-Media Thickness Partly Indicates the Prevention of Hypertension among Older Individuals in the General Population. Life (Basel) 2023; 13:1588. [PMID: 37511963 PMCID: PMC10381883 DOI: 10.3390/life13071588] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Structural atherosclerosis, as evaluated by carotid intima-media thickness (CIMT), is reported to be positively associated with hypertension. However, angiogenesis, which plays an important role in the progression of structural atherosclerosis, prevents hypertension by reducing peripheral vascular resistance. These associations evoke a contradiction: characteristics associated with the progression of structural atherosclerosis, which is related to hypertension, might prevent hypertension. To clarify novel mechanisms underlying the association between structural atherosclerosis and hypertension, multifaceted analyses are necessary. We performed several epidemiological studies based on this concept. This study summarizes those epidemiological studies and adds some discussion. Studies focusing on circulating CD34-positive cells, single-nucleotide polymorphisms (SNPs) of vascular endothelial growth factor (VEGF), SNPs in BRACA1-associated protein (BRAP), platelets, human T-cell leukemia virus type 1 (HTLV-1), and SNPs in aldehyde dehydrogenase 2 (ALDH2) have shown that active endothelial repair, which leads to the progression of structural atherosclerosis, helps prevent hypertension. These associations indicate that the progression of structural atherosclerosis could act as a marker of angiogenesis, which reduces peripheral vascular resistance. In general, a positive association between structural atherosclerosis and hypertension has been reported. However, the progression of structural atherosclerosis could act as a marker of activity that prevents hypertension via reductions in peripheral vascular resistance.
Collapse
Affiliation(s)
- Yuji Shimizu
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka 537-0025, Japan
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
33
|
Zhao N, Guo P, Tang M, Yang F, Zhang T, Mao R. Evidence for a causal relationship between psoriasis and cutaneous melanoma: a bidirectional two-sample Mendelian randomized study. Front Immunol 2023; 14:1201167. [PMID: 37503344 PMCID: PMC10368886 DOI: 10.3389/fimmu.2023.1201167] [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: 04/06/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Background and objective Existing cross-sectional and retrospective studies were unable to establish a causal relationship between psoriasis and cutaneous melanoma (CM). We sought to evaluate the causal role between psoriasis and CM. Methods We performed a bidirectional two-sample Mendelian randomization analysis using summary statistics from genome-wide association studies of psoriasis and CM among individuals of predominantly European ancestry. Mendelian randomization-Egger regression, inverse variance weighting, Mendelian Randomization Pleiotropy RESidual Sum and Outlier, weighted mode, and weighted median were used to examine the causal effect between psoriasis and CM. Results Genetically predicted psoriasis was a significant risk factor for CM (odds ratio, 1.69; 95% confidence interval, 1.15-2.48; P = 0.025). In contrast, no association was observed between genetically predicted CM and psoriasis. Conclusion Our findings corroborated the existence of genetically predicted psoriasis increases risk of CM. Enhanced early screening of cutaneous melanoma in patients with psoriasis may improve clinical burden. However, we did not find evidence for a causal link from CM to psoriasis, so further studies are required to elucidate the effect of CM activity on psoriasis.
Collapse
Affiliation(s)
- Nana Zhao
- Department of Operating Room, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Pengsen Guo
- The Center of Gastrointestinal and Minimally Invasive Surgery, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Mei Tang
- Department of Operating Room, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Fan Yang
- Emergency Department, Peking University Third Hospital, Peking University School of Medicine, Beijing, China
| | - Tongtong Zhang
- The Center of Gastrointestinal and Minimally Invasive Surgery, The Third People’s Hospital of Chengdu, Chengdu, China
- Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, Sichuan, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Han X, Li Q, Zhang S, Sun L, Liu W, Wang J. Inhibition of NEMO alleviates arthritis by blocking the M1 macrophage polarization. Int Immunopharmacol 2023; 117:109983. [PMID: 37012872 DOI: 10.1016/j.intimp.2023.109983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/15/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023]
Abstract
The nuclear factor-kappa B (NF-κB) signaling pathway and macrophages are critically involved in the pathogenesis of rheumatoid arthritis (RA). Recent studies have identified NF-κB essential modulator (NEMO), a regulatory subunit of the inhibitor of NF-κB kinase (IKK), as a potential target to inhibit NF-κB signaling pathway. Here, we investigated the interactions between NEMO and M1 macrophage polarization in RA. NEMO inhibition led to the suppression of proinflammatory cytokines secreted from M1 macrophages in collagen-induced arthritis mice. From lipopolysaccharide (LPS)-stimulated RAW264, knocking down NEMO blocked M1 macrophage polarization accompanied by lesser M1 proinflammatory subtype. Our findings link the novel regulatory component of NF-κB signaling and human arthritis pathologies which will pave the way towards the identification of new therapeutic targets and the development of innovative preventive strategies.
Collapse
|
35
|
Candellier A, Issa N, Grissi M, Brouette T, Avondo C, Gomila C, Blot G, Gubler B, Touati G, Bennis Y, Caus T, Brazier M, Choukroun G, Tribouilloy C, Kamel S, Boudot C, Hénaut L. Indoxyl-sulfate activation of the AhR- NF-κB pathway promotes interleukin-6 secretion and the subsequent osteogenic differentiation of human valvular interstitial cells from the aortic valve. J Mol Cell Cardiol 2023; 179:18-29. [PMID: 36967106 DOI: 10.1016/j.yjmcc.2023.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Calcific aortic stenosis (CAS) is more prevalent, occurs earlier, progresses faster and has worse outcomes in patients with chronic kidney disease (CKD). The uremic toxin indoxyl sulfate (IS) is powerful predictor of cardiovascular mortality in these patients and a strong promoter of ectopic calcification whose role in CAS remains poorly studied. The objective of this study was to evaluate whether IS influences the mineralization of primary human valvular interstitial cells (hVICs) from the aortic valve. METHODS Primary hVICs were exposed to increasing concentrations of IS in osteogenic medium (OM). The hVICs' osteogenic transition was monitored by qRT-PCRs for BMP2 and RUNX2 mRNA. Cell mineralization was assayed using the o-cresolphthalein complexone method. Inflammation was assessed by monitoring NF-κB activation using Western blots as well as IL-1β, IL-6 and TNF-α secretion by ELISAs. Small interfering RNA (siRNA) approaches enabled us to determine which signaling pathways were involved. RESULTS Indoxyl-sulfate increased OM-induced hVICs osteogenic transition and calcification in a concentration-dependent manner. This effect was blocked by silencing the receptor for IS (the aryl hydrocarbon receptor, AhR). Exposure to IS promoted p65 phosphorylation, the blockade of which inhibited IS-induced mineralization. Exposure to IS promoted IL-6 secretion by hVICs, a phenomenon blocked by silencing AhR or p65. Incubation with an anti-IL-6 antibody neutralized IS's pro-calcific effects. CONCLUSION IS promotes hVIC mineralization through AhR-dependent activation of the NF-κB pathway and the subsequent release of IL-6. Further research should seek to determine whether targeting inflammatory pathways can reduce the onset and progression of CKD-related CAS.
Collapse
Affiliation(s)
- Alexandre Candellier
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France; Department of Nephrology Dialysis and Transplantation, Amiens University Hospital, Amiens, France
| | - Nervana Issa
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Maria Grissi
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Théo Brouette
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Carine Avondo
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Cathy Gomila
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Gérémy Blot
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Brigitte Gubler
- Department of Immunology, Amiens University Hospital, Amiens, France; Department of Molecular Oncobiology, Amiens University Hospital, 80054, France; EA4666 - HEMATIM, CURS, Picardie Jules Verne University, Amiens 80054, France
| | - Gilles Touati
- Department of Cardiac Surgery, Amiens University Hospital, Amiens, France
| | - Youssef Bennis
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Thierry Caus
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France; Department of Cardiac Surgery, Amiens University Hospital, Amiens, France
| | - Michel Brazier
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France; Department of Biochemistry, Amiens University Hospital, Amiens, France
| | - Gabriel Choukroun
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France; Department of Nephrology Dialysis and Transplantation, Amiens University Hospital, Amiens, France
| | - Christophe Tribouilloy
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France; Department of Cardiology, Amiens University Hospital, Amiens, France
| | - Saïd Kamel
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France; Department of Biochemistry, Amiens University Hospital, Amiens, France
| | - Cédric Boudot
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France
| | - Lucie Hénaut
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Amiens, France.
| |
Collapse
|
36
|
Alam M, Hasan GM, Eldin SM, Adnan M, Riaz MB, Islam A, Khan I, Hassan MI. Investigating regulated signaling pathways in therapeutic targeting of non-small cell lung carcinoma. Biomed Pharmacother 2023; 161:114452. [PMID: 36878052 DOI: 10.1016/j.biopha.2023.114452] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is the most common malignancy worldwide. The signaling cascades are stimulated via genetic modifications in upstream signaling molecules, which affect apoptotic, proliferative, and differentiation pathways. Dysregulation of these signaling cascades causes cancer-initiating cell proliferation, cancer development, and drug resistance. Numerous efforts in the treatment of NSCLC have been undertaken in the past few decades, enhancing our understanding of the mechanisms of cancer development and moving forward to develop effective therapeutic approaches. Modifications of transcription factors and connected pathways are utilized to develop new treatment options for NSCLC. Developing designed inhibitors targeting specific cellular signaling pathways in tumor progression has been recommended for the therapeutic management of NSCLC. This comprehensive review provided deeper mechanistic insights into the molecular mechanism of action of various signaling molecules and their targeting in the clinical management of NSCLC.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Sayed M Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Muhammad Bilal Riaz
- Faculty of Applied Physics and Mathematics, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdnask, Poland; Department of Computer Science and Mathematics, Lebanese American University, Byblos, Lebanon
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
37
|
Tan Q, Yao CL. NEMO involves in NF-κB activation by interaction with p65 and promoting its nuclear translocation in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2023; 134:108637. [PMID: 36841517 DOI: 10.1016/j.fsi.2023.108637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
NEMO (nuclear factor-κB <NF-κB> essential modulator) plays an important role in activating NF-κB signaling pathway, p65 is a pivotal positive-regulator of NF-κB family. However, the role of NEMO in p65-triggered immune activation in teleost is largely unknown. In the present study, the cDNA sequence of LcNEMO was identified from the large yellow croaker (Larimichthys crocea). The predicated LcNEMO protein encoded 565 amino acids, consisting of a N-terminal NEMO domain, followed by two coiled coil (CC) motifs, a CC2-leucine zipper (CC2-LZ) domain, and a C-terminal zinc finger (ZnF) domain. Quantitative PCR showed that the strongest constitutive expression of LcNEMO was detected in blood and the inductive expression of it significantly enhanced after LPS and poly I:C challenge. The effect of LcNEMO on p65, RelB and cRel associated-immune activation detected by dual-luciferase reporter system assay indicated that Lcp65-triggered NF-κB, TNF-α and IL-1β activation could be significantly enhanced by LcNEMO. Furthermore, Co-IP revealed that the protein-protein interaction was existed between LcNEMO and Lcp65. Western blot and confocal microscope observation displayed that Lcp65 nuclear translocation could be promoted by LcNEMO with a dose- and time-dependent manner, which was further verified by RNA interference of LcNEMO expression. Our findings suggest that LcNEMO may be crucial in immune response by promoting p65-mediated immune activation.
Collapse
Affiliation(s)
- Qing Tan
- Fisheries College, Jimei University, Xiamen, 361021, PR China
| | - Cui-Luan Yao
- Fisheries College, Jimei University, Xiamen, 361021, PR China.
| |
Collapse
|
38
|
Analysis of NFKB1 and NFKB2 gene expression in the blood of patients with sudden sensorineural hearing loss. Int J Pediatr Otorhinolaryngol 2023; 166:111470. [PMID: 36773447 DOI: 10.1016/j.ijporl.2023.111470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Sudden Sensorineural Hearing Loss (SSNHL) is an increasingly common health problem today. Although the direct mortality rate of this disorder is relatively low, its impact on quality of life is enormous; this is why accurate identification of pathogenesis and influencing factors in the disease process can play an essential role in preventing and treating the disease. Acute inflammation, which leads to chronic inflammation due to aberrant expression of inflammation-mediating genes, may play a significant role in the pathogenesis of the disease. The essential Nuclear factor kappa B (NF-kB) pathway genes, NFKB1 and NFKB2, serve as prothrombotic agents when expressed abnormally, compromising the cochlea by disrupting the endolymphatic potential and causing SSNHL. METHODS This study investigates the expression levels of NFKB1 and NFKB2 in peripheral blood (PB) through a quantitative polymerase chain reaction in 50 Iranian patients with SSNHL, and 50 healthy volunteers were of the same age and sex as controls. RESULTS As a result, NFKB2 expression levels in patients were higher than in controls, regardless of sex or age (posterior beta = 0.619, adjusted P-value = 0.016), and NFKB1 expression levels did not show significant differences between patients and controls. The expression levels of NFKB1 and NFKB2 had significantly strong positive correlations in both SSNHL patients and healthy individuals (r = 0.620, P = 0.001 and r = 0.657, P 0.001, respectively), suggesting the presence of an interconnected network. CONCLUSION NFKB2 has been identified as a significant inflammatory factor in the pathophysiology of SSNHL disease. Inflammation can play an essential role in developing SSNHL, and our findings could be used as a guide for future research.
Collapse
|
39
|
Olędzka AJ, Czerwińska ME. Role of Plant-Derived Compounds in the Molecular Pathways Related to Inflammation. Int J Mol Sci 2023; 24:ijms24054666. [PMID: 36902097 PMCID: PMC10003729 DOI: 10.3390/ijms24054666] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Inflammation is the primary response to infection and injury. Its beneficial effect is an immediate resolution of the pathophysiological event. However, sustained production of inflammatory mediators such as reactive oxygen species and cytokines may cause alterations in DNA integrity and lead to malignant cell transformation and cancer. More attention has recently been paid to pyroptosis, which is an inflammatory necrosis that activates inflammasomes and the secretion of cytokines. Taking into consideration that phenolic compounds are widely available in diet and medicinal plants, their role in the prevention and support of the treatment of chronic diseases is apparent. Recently, much attention has been paid to explaining the significance of isolated compounds in the molecular pathways related to inflammation. Therefore, this review aimed to screen reports concerning the molecular mode of action assigned to phenolic compounds. The most representative compounds from the classes of flavonoids, tannins, phenolic acids, and phenolic glycosides were selected for this review. Our attention was focused mainly on nuclear factor-κB (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), and mitogen-activated protein kinase (MAPK) signaling pathways. Literature searching was performed using Scopus, PubMed, and Medline databases. In conclusion, based on the available literature, phenolic compounds regulate NF-κB, Nrf2, and MAPK signaling, which supports their potential role in chronic inflammatory disorders, including osteoarthritis, neurodegenerative diseases, cardiovascular, and pulmonary disorders.
Collapse
Affiliation(s)
- Agata J. Olędzka
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, 1B Banacha Str., 02-097 Warsaw, Poland
| | - Monika E. Czerwińska
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, 1B Banacha Str., 02-097 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-116-61-85
| |
Collapse
|
40
|
Zhao M, Wang X, Kumar SA, Yao Y, Sun M. A Pharmacological Insight of Piperlongumine, Bioactive Validating Its Therapeutic Efficacy as a Drug to Treat Inflammatory Diseases. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2023. [DOI: 10.1134/s1068162023020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
41
|
Bioactivity, Molecular Mechanism, and Targeted Delivery of Flavonoids for Bone Loss. Nutrients 2023; 15:nu15040919. [PMID: 36839278 PMCID: PMC9960663 DOI: 10.3390/nu15040919] [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: 01/11/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Skeletal disabilities are a prominent burden on the present population with an increasing life span. Advances in osteopathy have provided various medical support for bone-related diseases, including pharmacological and prosthesis interventions. However, therapeutics and post-surgery complications are often reported due to side effects associated with modern-day therapies. Thus, therapies utilizing natural means with fewer toxic or other side effects are the key to acceptable interventions. Flavonoids constitute a class of bioactive compounds found in dietary supplements, and their pharmacological attributes have been well appreciated. Recently, flavonoids' role is gaining renowned interest for its effect on bone remodeling. A wide range of flavonoids has been found to play a pivotal role in the major bone signaling pathways, such as wingless-related integration site (Wnt)/β-catenin, bone morphogenetic protein (BMP)/transforming growth factor (TGF)-β, mitogen-activated protein kinase (MAPK), etc. However, the reduced bioavailability and the absorption of flavonoids are the major limitations inhibiting their use against bone-related complications. Recent utilization of nanotechnological approaches and other delivery methods (biomaterial scaffolds, micelles) to target and control release can enhance the absorption and bioavailability of flavonoids. Thus, we have tried to recapitulate the understanding of the role of flavonoids in regulating signaling mechanisms affecting bone remodeling and various delivery methods utilized to enhance their therapeutical potential in treating bone loss.
Collapse
|
42
|
Rashid MU, Lorzadeh S, Gao A, Ghavami S, Coombs KM. PSMA2 knockdown impacts expression of proteins involved in immune and cellular stress responses in human lung cells. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166617. [PMID: 36481484 DOI: 10.1016/j.bbadis.2022.166617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Proteasome subunit alpha type-2 (PSMA2) is a critical component of the 20S proteasome, which is the core particle of the 26S proteasome complex and is involved in cellular protein quality control by recognizing and recycling defective proteins. PSMA2 expression dysregulation has been detected in different human diseases and viral infections. No study yet has reported PSMA2 knockdown (KD) effects on the cellular proteome. METHODS We used SOMAScan, an aptamer-based multiplexed technique, to measure >1300 human proteins to determine the impact of PSMA2 KD on A549 human lung epithelial cells. RESULTS PSMA2 KD resulted in significant dysregulation of 52 cellular proteins involved in different bio-functions, including cellular movement and development, cell death and survival, and cancer. The immune system and signal transduction were the most affected cellular functions. PSMA2 KD caused dysregulation of several signaling pathways involved in immune response, cytokine signaling, organismal growth and development, cellular stress and injury (including autophagy and unfolded protein response), and cancer responses. CONCLUSIONS In summary, this study helps us better understand the importance of PSMA2 in different cellular functions, signaling pathways, and human diseases.
Collapse
Affiliation(s)
- Mahamud-Ur Rashid
- University of Manitoba, Department of Medical Microbiology & Infectious Diseases, Room 543 Basic Medical Sciences Building, 745 Bannatyne Ave., Winnipeg, MB R3E 0J9, Canada; Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Ang Gao
- Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Kevin M Coombs
- University of Manitoba, Department of Medical Microbiology & Infectious Diseases, Room 543 Basic Medical Sciences Building, 745 Bannatyne Ave., Winnipeg, MB R3E 0J9, Canada; Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada; Children's Hospital Research Institute of Manitoba, Room 513, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
43
|
Bradfield CJ, Liang JJ, Ernst O, John SP, Sun J, Ganesan S, de Jesus AA, Bryant CE, Goldbach-Mansky R, Fraser IDC. Biphasic JNK signaling reveals distinct MAP3K complexes licensing inflammasome formation and pyroptosis. Cell Death Differ 2023; 30:589-604. [PMID: 36624264 PMCID: PMC9950443 DOI: 10.1038/s41418-022-01106-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023] Open
Abstract
Kinase signaling in the tiered activation of inflammasomes and associated pyroptosis is a prime therapeutic target for inflammatory diseases. While MAPKs subsume pivotal roles during inflammasome priming, specifically the MAP3K7/JNK1/NLRP3 licensing axis, their involvement in successive steps of inflammasome activation is poorly defined. Using live-cell MAPK biosensors to focus on the inflammasome triggering event allowed us to identify a subsequent process of biphasic JNK activation. We find that this biphasic post-trigger JNK signaling initially facilitates the mitochondrial reactive oxygen species generation needed to support core inflammasome formation, then supports the gasdermin-mediated cell permeation required for release of active IL-1β from human macrophages. We further identify and characterize a xanthine oxidase-ROS activated MAP3K5/JNK2 substrate licensing complex as a novel regulator of the GSDMD mobilization which precedes pyroptosis. We show that inhibitors targeting this MAP3K5 cascade alleviate morbidity in mouse models of colitis and dampen both augmented IL-1β release and cell permeation in monocytes derived from patients with gain-of-function inflammasomopathies.
Collapse
Affiliation(s)
- Clinton J Bradfield
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Jonathan J Liang
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
- University of Cambridge, Department of Medicine, Cambridge, UK
| | - Orna Ernst
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Sinu P John
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | - Jing Sun
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA
| | | | - Adriana A de Jesus
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, NIAID, Bethesda, MD, USA
| | - Clare E Bryant
- University of Cambridge, Department of Medicine, Cambridge, UK
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, NIAID, Bethesda, MD, USA
| | - Iain D C Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, NIAID, Bethesda, MD, USA.
| |
Collapse
|
44
|
Huang JY, Ma KSK, Wang LT, Chiang CH, Yang SF, Wang CH, Wang PH. The Risk of Endometrial Cancer and Uterine Sarcoma Following Endometriosis or Pelvic Inflammatory Disease. Cancers (Basel) 2023; 15:cancers15030833. [PMID: 36765791 PMCID: PMC9913480 DOI: 10.3390/cancers15030833] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 01/31/2023] Open
Abstract
The relationship between uterine corpus cancer and endometriosis was conflicting. We aimed to determine the risk of uterine cancer in patients with endometriosis or pelvic inflammatory disease (PID). In this population-based cohort study, a total of 135,236 females with endometriosis (n = 20,510) or PID (n = 114,726), as well as 135,236 age-matched controls, were included. Cox regression models estimated the risk of uterine cancer in each group. Sub-outcomes of risk for uterine corpus cancer included endometrial cancer and uterine sarcoma were analyzed. An age subgroup analysis was performed to determine the moderator effect of age. A landmark analysis depicted the time varying effect of endometriosis and PID. A propensity score matching analysis was conducted to validate the findings. Patients with endometriosis had significantly higher risk of endometrial cancer (adjusted hazard ratio, aHR = 2.92; 95% CI = 2.12-4.03) and uterine sarcoma (aHR = 5.83; 95% CI = 2.02-16.89), while PID was not associated with the risk of uterine cancer. The increased risk of uterine cancer in patients with endometriosis persisted after propensity score matching (aHR = 2.83, 95%CI = 1.70-4.71). The greatest risk of endometrial cancer occurred in patients who had endometriosis for 37 to 60 months (adjusted relative risk, aRR = 9.15, 95% CI = 4.40-19.02). Females aged 12 to 35 years were at the greatest risk of endometriosis-associated uterine cancer (RR = 6.97, 95% CI = 3.41-14.26). In conclusion, patients with endometriosis were at great risk of uterine cancer, including endometrial cancer and uterine sarcoma, compared with propensity score-matched populations and compared with patients of PID. Younger females with endometriosis and patients who had endometriosis for three to five years were at the greatest risk of endometriosis-associated uterine cancer.
Collapse
Affiliation(s)
- Jing-Yang Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Kevin Sheng-Kai Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Dermatology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Orthodontics and Dentofacial Orthopedics, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118, USA
| | - Li-Tzu Wang
- Department of Obstetrics & Gynecology, National Taiwan University Hospital & College of Medicine, Taipei 11031, Taiwan
| | - Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Cambridge, MA 02138, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Chun-Hao Wang
- Department of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Po-Hui Wang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: ; Tel.: +886-4-24739595 (ext. 21721); Fax: +886-4-24738493
| |
Collapse
|
45
|
Huang ZQ, Luo W, Li WX, Chen P, Wang Z, Chen RJ, Wang Y, Huang WJ, Liang G. Costunolide alleviates atherosclerosis in high-fat diet-fed ApoE -/- mice through covalently binding to IKKβ and inhibiting NF-κB-mediated inflammation. Acta Pharmacol Sin 2023; 44:58-70. [PMID: 35710877 PMCID: PMC9813247 DOI: 10.1038/s41401-022-00928-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/26/2022] [Indexed: 01/18/2023]
Abstract
Costunolide (CTD) is a sesquiterpene lactone isolated from costus root and exhibits various biological activities including anti-inflammation. Since atherosclerosis is a chronic inflammatory disease, we herein investigated the anti-atherosclerotic effects of CTD and the underlying mechanism. Atherosclerosis was induced in ApoE-/- mice by feeding them with a high-fat diet (HFD) for 8 weeks, followed by administration of CTD (10, 20 mg ·kg-1·d-1, i.g.) for 8 weeks. We showed that CTD administration dose-dependently alleviated atherosclerosis in HFD-fed ApoE-/- mice. Furthermore, we found that CTD dose-dependently reduced inflammatory responses in aortas of the mice, as CTD prevented infiltration of inflammatory cells in aortas and attenuated oxLDL uptake in macrophages, leading to reduced expression of pro-inflammatory and pro-fibrotic molecules in aortas. Similar results were observed in oxLDL-stimulated mouse primary peritoneal macrophages (MPMs) in vitro. We showed that pretreatment with CTD (2.5, 5. 10 μM) restrained oxLDL-induced inflammatory responses in MPMs by blocking pro-inflammatory NF-κB/p65 signaling pathway. We further demonstrated that CTD inactivated NF-κB via covalent binding to cysteine 179 on IKKβ, a canonical upstream regulator of NF-κB, reducing its phosphorylation and leading to conformational change in the active loop of IKKβ. Our results discover IKKβ as the target of CTD for its anti-inflammatory activity and elucidate a molecular mechanism underlying the anti-atherosclerosis effect of CTD. CTD is a potentially therapeutic candidate for retarding inflammatory atherosclerotic diseases.
Collapse
Affiliation(s)
- Zhu-Qi Huang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Xin Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Pan Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhe Wang
- Department of Pharmacy, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Rui-Jie Chen
- Department of Pharmacy, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Jian Huang
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China.
- Department of Cardiology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
46
|
Wu Y, Yang Y, Wang L, Chen Y, Han X, Sun L, Chen H, Chen Q. Effect of Bifidobacterium on osteoclasts: TNF-α/NF-κB inflammatory signal pathway-mediated mechanism. Front Endocrinol (Lausanne) 2023; 14:1109296. [PMID: 36967748 PMCID: PMC10034056 DOI: 10.3389/fendo.2023.1109296] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
Osteoporosis is a systemic multifactorial bone disease characterized by low bone quality and density and bone microstructure damage, increasing bone fragility and fracture vulnerability. Increased osteoclast differentiation and activity are important factors contributing to bone loss, which is a common pathological manifestation of bone diseases such as osteoporosis. TNF-a/NF-κB is an inflammatory signaling pathway with a key regulatory role in regulating osteoclast formation, and the classical pathway RANKL/RANK/OPG assists osteoclast formation. Activation of this inflammatory pathway promotes the formation of osteoclasts and accelerates the process of osteoporosis. Recent studies and emerging evidence have consistently demonstrated the potential of probiotics to modulate bone health. Secretions of Bifidobacterium, a genus of probiotic bacteria in the phylum Actinobacteria, such as short-chain fatty acids, equol, and exopolysaccharides, have indicated beneficial effects on bone health. This review discusses the molecular mechanisms of the TNF-a/NF-κB inflammatory pathway in regulating osteoclast formation and describes the secretions produced by Bifidobacterium and their potential effects on bone health through this pathway, opening up new directions for future research.
Collapse
Affiliation(s)
- Yue Wu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunjiao Yang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lan Wang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiding Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuke Han
- College of Acupuncture & Tuina, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lisha Sun
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huizhen Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Qiu Chen,
| |
Collapse
|
47
|
Kurniati D, Hirai S, Egashira Y. Effect of apigenin on tryptophan metabolic key enzymes expression in lipopolysaccharide-induced microglial cells and its mechanism. Heliyon 2022; 9:e12743. [PMID: 36685364 PMCID: PMC9852672 DOI: 10.1016/j.heliyon.2022.e12743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/09/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
[Aims] Flavonoid apigenin (API) has a wide range of biological functions, particularly anti-inflammation. Indoleamine 2,3-dioxygenase (IDO) and 2-Amino-3-carboxymuconate-6-semialdehyde decarboxylase (ACMSD) are important tryptophan metabolic enzymes that play pivotal roles in the production of toxic metabolite quinolinic acid. However, the relationship between inflammation and ACMSD remains unclear. The present study investigated the relationship between inflammation and tryptophan metabolic key enzymes. Similarly, the anti-inflammatory effect of API on important tryptophan metabolic enzymes was examined in lipopolysaccharide (LPS)-treated microglial cells. [Main methods] MG6 cells were exposed to LPS with or without API treatment for 24-48 h. IDO and ACMSD mRNA expression and production of inflammatory mediators were analyzed. Activation of inflammatory signaling pathways, such as mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB), was also examined to study the mechanism of API in the inflammatory state. [Key findings] LPS suppressed ACMSD expression and enhanced IDO expression. However, API elevated ACMSD mRNA expression and suppressed IDO mRNA expression in LPS-treated MG6 cells. Furthermore, API suppressed interleukin-6 and nitric oxide production, whereas overproduction of inflammatory mediators enhanced IDO expression and assisted tryptophan degradation. API also inhibited activation of extracellular signal-regulated kinase (Erk) and jun N-terminal kinase (JNK) MAPK, and degradation of IκBα. [Significance] These results indicate alteration of ACMSD expression under inflammatory conditions. Moreover, API recovers expression of tryptophan metabolic key enzymes, which may be mediated by inhibition of proinflammatory mediator production via inactivation of Erk, JNK MAPK, and NF-κB pathways in LPS-stimulated microglial cells.
Collapse
Affiliation(s)
- Dian Kurniati
- Laboratory of Food and Nutrition, Division of Applied Biochemistry, Graduate School of Horticulture, Chiba University, 648, Matsudo, Matsudo-shi, Chiba, 271-8510, Japan,Department of Food Technology, Faculty of Agricultural Industrial Technology, Universitas Padjadjaran, Sumedang KM. 21, Jatinangor, 40600, West Java, Indonesia
| | - Shizuka Hirai
- Laboratory of Food and Nutrition, Division of Applied Biochemistry, Graduate School of Horticulture, Chiba University, 648, Matsudo, Matsudo-shi, Chiba, 271-8510, Japan
| | - Yukari Egashira
- Laboratory of Food and Nutrition, Division of Applied Biochemistry, Graduate School of Horticulture, Chiba University, 648, Matsudo, Matsudo-shi, Chiba, 271-8510, Japan,Corresponding author.
| |
Collapse
|
48
|
Ismael S, Patrick D, Salman M, Parveen A, Stanfill AG, Ishrat T. Verapamil inhibits TXNIP-NLRP3 inflammasome activation and preserves functional recovery after intracerebral hemorrhage in mice. Neurochem Int 2022; 161:105423. [PMID: 36244583 DOI: 10.1016/j.neuint.2022.105423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/08/2022]
Abstract
Intracerebral hemorrhage (ICH) is the second most common type of stroke with no satisfactory treatment. Recent studies from our group and others indicated a potential positive effect of verapamil, a commonly prescribed calcium channel blocker, with thioredoxin-interacting protein (TXNIP) inhibitor properties, in ischemic stroke and cognitive disorders. It is unclear whether there would be a beneficial effect of verapamil administration in ICH. Therefore, this study was designed to determine the neuroprotective effects of verapamil in a murine ICH model. ICH was induced by stereotactic injection of collagenase type VII (0.075 U) into the right striatum of adult male C57BL/6 mice. Verapamil (0.15 mg/kg) or saline was administered intravenously at 1 h post-ICH followed by oral (1 mg/kg/d) administration in drinking water for 28 days. Motor and cognitive function were assessed using established tests for motor coordination, spatial learning, short- and long-term memory. A subset of animals was sacrificed at 72 h after ICH for molecular analysis. Verapamil treatment reduced expression of TXNIP and NOD-like receptor pyrin domain-containing-3 inflammasome activation in the perihematomal area. These protective effects of verapamil were associated with decreased proinflammatory mediators, microglial activation, and blood-brain barrier permeability markers and paralleled less phosphorylated nuclear factor kappa B level. Our findings also demonstrate that long-term low-dose verapamil effectively attenuated motor and cognitive impairments. Taken together, these data indicate that verapamil has therapeutic potential in improving acute motor function after ICH. Further investigations are needed to confirm whether verapamil treatment could be a promising candidate for clinical trials.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Devlin Patrick
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA; Department of Acute and Tertiary Care, College of Nursing, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mohd Salman
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Arshi Parveen
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ansley Grimes Stanfill
- Department of Acute and Tertiary Care, College of Nursing, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
49
|
Sadiku OO, Rodríguez-Seijo A. Metabolic and genetic derangement: a review of mechanisms involved in arsenic and lead toxicity and genotoxicity. Arh Hig Rada Toksikol 2022; 73:244-255. [PMID: 36607725 PMCID: PMC9985351 DOI: 10.2478/aiht-2022-73-3669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/01/2022] [Accepted: 10/01/2022] [Indexed: 01/07/2023] Open
Abstract
Urbanisation and industrialisation are on the rise all over the world. Environmental contaminants such as potentially toxic elements (PTEs) are directly linked with both phenomena. Two PTEs that raise greatest concern are arsenic (As) and lead (Pb) as soil and drinking water contaminants, whether they are naturally occurring or the consequence of human activities. Both elements are potential carcinogens. This paper reviews the mechanisms by which As and Pb impair metabolic processes and cause genetic damage in humans. Despite efforts to ban or limit their use, due to high persistence both continue to pose a risk to human health, which justifies the need for further toxicological research.
Collapse
Affiliation(s)
- Olubusayo Olujimi Sadiku
- University of Lagos, College of Medicine, Faculty of Basic Medical Sciences, Department of Medical Laboratory Science, Lagos, Nigeria
| | - Andrés Rodríguez-Seijo
- University of Porto, Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Matosinhos, Portugal
- University of Porto, Faculty of Sciences, Biology Department, Porto, Portugal
- University of Vigo, Department of Plant Biology and Soil Sciences, Ourense, Spain
| |
Collapse
|
50
|
Engineered nanoparticles as emerging gene/drug delivery systems targeting the nuclear factor-κB protein and related signaling pathways in cancer. Biomed Pharmacother 2022; 156:113932. [DOI: 10.1016/j.biopha.2022.113932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
|