51
|
Qu JY, Xie HT, Xiao YT, Zhang YY, Hu ZX, Wang JS, Zhang MC, Xi H. The inhibition of p38 MAPK blocked inflammation to restore the functions of rat meibomian gland epithelial cells. Exp Eye Res 2023; 231:109470. [PMID: 37059216 DOI: 10.1016/j.exer.2023.109470] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/03/2023] [Accepted: 04/08/2023] [Indexed: 04/16/2023]
Abstract
Meibomian glands (MGs) are vital for ocular surface health. However, the roles of inflammation in the progression of meibomian gland dysfunction (MGD) are largely unknown. In this study, the roles of the inflammation factor interleukin-1β (IL-1β) via the p38 mitogen-activated protein kinases (MAPK) signaling pathway on rat meibomian gland epithelial cells (RMGECs) were explored. Eyelids from adult rat mice at 2 months and 2 years of age were stained with specific antibodies against IL-1β to identify inflammation levels. RMGECs were exposed to IL-1β and/or SB203580, a specific inhibitor of p38 MAPK signaling pathway, for 3 days. Cell proliferation, keratinization, lipid accumulation, and matrix metalloproteinases 9 (MMP9) expression were evaluated by MTT assay, polymerase chain reaction (PCR), immunofluorescence staining, apoptosis assay, lipid staining, and Western blot analyses. We found that IL-1β was significantly higher in the terminal ducts of MGs in rats with age-related MGD than in young rats. IL-1β inhibited cell proliferation, suppressed lipid accumulation and peroxisome proliferator activator receptor γ (PPARγ) expression, and promoted apoptosis while activating the p38 MAPK signaling pathway. Cytokeratin 1 (CK1), a marker for complete keratinization, and MMP9 in RMGECs were also up-regulated by IL-1β. SB203580 effectively diminished the effects of IL-1β on differentiation, keratinization, and MMP9 expression by blocking IL-1β-induced p38 MAPK activation, although it also inhibited cell proliferation. The inhibition of the p38 MAPK signaling pathway blocked IL-1β-induced differentiation reduction, hyperkeratinization, and MMP9 overexpression of RMGECs, which provides a potential therapy for MGD.
Collapse
Affiliation(s)
- Jing-Yu Qu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266000, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266000, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu-Ting Xiao
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ying-Ying Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhi-Xin Hu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia-Song Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Han Xi
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
52
|
Xiong D, Gao F, Shao J, Pan Y, Wang S, Wei D, Ye S, Chen Y, Chen R, Yue B, Li J, Chen J. Arctiin-encapsulated DSPE-PEG bubble-like nanoparticles inhibit alveolar epithelial type 2 cell senescence to alleviate pulmonary fibrosis via the p38/p53/p21 pathway. Front Pharmacol 2023; 14:1141800. [PMID: 36998607 PMCID: PMC10043219 DOI: 10.3389/fphar.2023.1141800] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/28/2023] [Indexed: 03/15/2023] Open
Abstract
Background: Idiopathic pulmonary fibrosis is a severe and deadly form of diffuse parenchymal lung disease and treatment options are few. Alveolar epithelial type 2 (AEC2) cell senescence is implicated in the pathogenies of IPF. A major bioactive compound from the traditional Chinese medicine Fructus arctii, arctiin (ARC) has robust anti-inflammatory, anti-senescence, and anti-fibrosis functions. However, the potential therapeutic effects of ARC on IPF and the underlying mechanisms involved are still unknown.Methods: First of all, ARC was identified as an active ingredient by network pharmacology analysis and enrichment analysis of F. arctii in treating IPF. We developed ARC-encapsulated DSPE-PEG bubble-like nanoparticles (ARC@DPBNPs) to increase ARC hydrophilicity and achieve high pulmonary delivery efficiency. C57BL/6 mice were used to establish a bleomycin (BLM)-induced pulmonary fibrosis model for assessing the treatment effect of ARC@DPBNPs on lung fibrosis and the anti-senescence properties of AEC2. Meanwhile, p38/p53 signaling in AEC2 was detected in IPF lungs, BLM-induced mice, and an A549 senescence model. The effects of ARC@DPBNPs on p38/p53/p21 were assessed in vivo and in vitro.Results: Pulmonary route of administration of ARC@DPBNPs protected mice against BLM-induced pulmonary fibrosis without causing significant damage to the heart, liver, spleen, or kidney. ARC@DPBNPs blocked BLM-induced AEC2 senescence in vivo and in vitro. The p38/p53/p21 signaling axis was significantly activated in the lung tissues of patients with IPF, senescent AEC2, and BLM-induced lung fibrosis. ARC@DPBNPs attenuated AEC2 senescence and pulmonary fibrosis by inhibiting the p38/p53/p21 pathway.Conclusion: Our data suggest that the p38/p53/p21 signaling axis plays a pivotal role in AEC2 senescence in pulmonary fibrosis. The p38/p53/p21 signaling axis inhibition by ARC@DPBNPs provides an innovative approach to treating pulmonary fibrosis in clinical settings.
Collapse
Affiliation(s)
- Dian Xiong
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Fei Gao
- Department of Emergency, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
- Department of Emergency, Nanjing General Hospital of Nanjing Military Region, Nanjing, China
| | - Jingbo Shao
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Yueyun Pan
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Song Wang
- Department of Intensive Care Medicine, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Dong Wei
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Shugao Ye
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Yuan Chen
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
| | - Rui Chen
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Bingqing Yue
- Department of Lung Transplantation, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Li
- Department of Chemistry, Fudan University, Shanghai, China
- *Correspondence: Jingyu Chen, ; Juan Li,
| | - Jingyu Chen
- Lung Transplantation Center, Department of Thoracic Surgery, Nanjing Medical University Affiliated Wuxi People’s Hospital, Wuxi, China
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- *Correspondence: Jingyu Chen, ; Juan Li,
| |
Collapse
|
53
|
Nalinratana N, Suriya U, Laprasert C, Wisidsri N, Poldorn P, Rungrotmongkol T, Limpanasitthikul W, Wu HC, Chang HS, Chansriniyom C. In vitro and in silico studies of 7'',8''-buddlenol D anti-inflammatory lignans from Carallia brachiata as p38 MAP kinase inhibitors. Sci Rep 2023; 13:3558. [PMID: 36864126 PMCID: PMC9981598 DOI: 10.1038/s41598-023-30475-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Excessive macrophage activation induces the release of high levels of inflammatory mediators which not only amplify chronic inflammation and degenerative diseases but also exacerbate fever and retard wound healing. To identify anti-inflammatory molecules, we examined Carallia brachiata-a medicinal terrestrial plant from Rhizophoraceae. Furofuran lignans [(-)-(7''R,8''S)-buddlenol D (1) and (-)-(7''S,8''S)-buddlenol D (2)] isolated from the stem and bark inhibited nitric oxide (half maximal inhibitory concentration (IC50): 9.25 ± 2.69 and 8.43 ± 1.20 micromolar for 1 and 2, respectively) and prostaglandin E2 (IC50: 6.15 ± 0.39 and 5.70 ± 0.97 micromolar for 1 and 2, respectively) productions in lipopolysaccharide-induced RAW264.7 cells. From western blotting, 1 and 2 suppressed LPS-induced inducible nitric oxide synthase and cyclooxygenase-2 expression in a dose-dependent manner (0.3-30 micromolar). Moreover, analysis of the mitogen-activated protein kinase (MAPK) signaling pathway showed decreased p38 phosphorylation levels in 1- and 2-treated cells, while phosphorylated ERK1/2 and JNK levels were unaffected. This discovery agreed with in silico studies which suggested 1 and 2 bound to the ATP-binding site in p38-alpha MAPK based on predicted binding affinity and intermolecular interaction docking. In summary, 7'',8''-buddlenol D epimers demonstrated anti-inflammatory activities via p38 MAPK inhibition and may be used as viable anti-inflammatory therapies.
Collapse
Affiliation(s)
- Nonthaneth Nalinratana
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Utid Suriya
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chanyanuch Laprasert
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nakuntwalai Wisidsri
- Faculty of Integrative Medicine, Rajamangala University of Technology Thanyaburi, Pathum Thani, 12130, Thailand
| | - Preeyaporn Poldorn
- Biocatalyst and Environmental Biotechnology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanyada Rungrotmongkol
- Biocatalyst and Environmental Biotechnology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Ho-Cheng Wu
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Hsun-Shuo Chang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chaisak Chansriniyom
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Natural Products and Nanoparticles Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
54
|
Atanasova VS, de Jesus Cardona C, Hejret V, Tiefenbacher A, Mair T, Tran L, Pfneissl J, Draganić K, Binder C, Kabiljo J, Clement J, Woeran K, Neudert B, Wohlhaupter S, Haase A, Domazet S, Hengstschläger M, Mitterhauser M, Müllauer L, Tichý B, Bergmann M, Schweikert G, Hartl M, Dolznig H, Egger G. Mimicking Tumor Cell Heterogeneity of Colorectal Cancer in a Patient-derived Organoid-Fibroblast Model. Cell Mol Gastroenterol Hepatol 2023; 15:1391-1419. [PMID: 36868311 PMCID: PMC10141529 DOI: 10.1016/j.jcmgh.2023.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND & AIMS Patient-derived organoid cancer models are generated from epithelial tumor cells and reflect tumor characteristics. However, they lack the complexity of the tumor microenvironment, which is a key driver of tumorigenesis and therapy response. Here, we developed a colorectal cancer organoid model that incorporates matched epithelial cells and stromal fibroblasts. METHODS Primary fibroblasts and tumor cells were isolated from colorectal cancer specimens. Fibroblasts were characterized for their proteome, secretome, and gene expression signatures. Fibroblast/organoid co-cultures were analyzed by immunohistochemistry and compared with their tissue of origin, as well as on gene expression levels compared with standard organoid models. Bioinformatics deconvolution was used to calculate cellular proportions of cell subsets in organoids based on single-cell RNA sequencing data. RESULTS Normal primary fibroblasts, isolated from tumor adjacent tissue, and cancer associated fibroblasts retained their molecular characteristics in vitro, including higher motility of cancer associated compared with normal fibroblasts. Importantly, both cancer-associated fibroblasts and normal fibroblasts supported cancer cell proliferation in 3D co-cultures, without the addition of classical niche factors. Organoids grown together with fibroblasts displayed a larger cellular heterogeneity of tumor cells compared with mono-cultures and closely resembled the in vivo tumor morphology. Additionally, we observed a mutual crosstalk between tumor cells and fibroblasts in the co-cultures. This was manifested by considerably deregulated pathways such as cell-cell communication and extracellular matrix remodeling in the organoids. Thrombospondin-1 was identified as a critical factor for fibroblast invasiveness. CONCLUSION We developed a physiological tumor/stroma model, which will be vital as a personalized tumor model to study disease mechanisms and therapy response in colorectal cancer.
Collapse
Affiliation(s)
- Velina S Atanasova
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | | | - Václav Hejret
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Andreas Tiefenbacher
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Kristina Draganić
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Carina Binder
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Julijan Kabiljo
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Clinic of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Janik Clement
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Katharina Woeran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Barbara Neudert
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Astrid Haase
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sandra Domazet
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | | | | | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Boris Tichý
- CEITEC-Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michael Bergmann
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Clinic of General Surgery, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schweikert
- Max Planck Institute for Intelligent Systems, Tübingen, Germany; Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Markus Hartl
- Department of Biochemistry and Cell Biology, Max Perutz Labs, Vienna BioCenter (VBC), University of Vienna, Vienna, Austria; Mass Spectrometry Facility, Max Perutz Labs, Vienna BioCenter, University of Vienna, Vienna, Austria
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria.
| | - Gerda Egger
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
55
|
Luteoloside Induces G0/G1 Phase Arrest of Neuroblastoma Cells by Targeting p38 MAPK. Molecules 2023; 28:molecules28041748. [PMID: 36838737 PMCID: PMC9966487 DOI: 10.3390/molecules28041748] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Luteoloside has shown anti-inflammatory, antiviral, and antitumor properties. However, the effect and mechanism of luteoloside on neuroblastoma cells remain unknown. The proliferation of human neuroblastoma cells (SH-SY5Y and SK-N-AS) treated with different concentrations of luteoloside (0, 12.5, 25, and 50 μM) was detected by the MTT assay and colony formation assay. Cell apoptosis and cell cycle were examined by Hoechst staining and flow cytometry. A subcutaneous tumorigenesis model was established in nude mice to evaluate the effect of luteoloside on tumor growth in vivo. Bioinformatics, molecular docking techniques, and cellular thermal shift assays were utilized to predict the potential targets of luteoloside in neuroblastoma. The p38 MAPK inhibitor SB203580 was used to confirm the role of p38 MAPK. Luteoloside inhibited the proliferation of neuroblastoma cells in vitro and in vivo. Luteoloside slightly induced cellular G0/G1 phase arrest and reduced the expression levels of G0/G1 phase-related genes and the proteins cyclin D1, CDK4, and C-myc, which are downregulated by p38 MAPK pathways. Meanwhile, p38 was identified as the target of luteoloside, and inhibition of p38 MAPK reversed the inhibitory effect of luteoloside on neuroblastoma cells. Luteoloside is a potential anticancer drug for treating neuroblastoma by activating p38 MAPK.
Collapse
|
56
|
Gabrielli AP, Weidling I, Ranjan A, Wang X, Novikova L, Chowdhury SR, Menta B, Berkowicz A, Wilkins HM, Peterson KR, Swerdlow RH. Mitochondria Profoundly Influence Apolipoprotein E Biology. J Alzheimers Dis 2023; 92:591-604. [PMID: 36776072 DOI: 10.3233/jad-221177] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Mitochondria can trigger Alzheimer's disease (AD)-associated molecular phenomena, but how mitochondria impact apolipoprotein E (APOE; apoE) is not well known. OBJECTIVE Consider whether and how mitochondrial biology influences APOE and apoE biology. METHODS We measured APOE expression in human SH-SY5Y neuronal cells with different forms of mitochondrial dysfunction including total, chronic mitochondrial DNA (mtDNA) depletion (ρ0 cells); acute, partial mtDNA depletion; and toxin-induced mitochondrial dysfunction. We further assessed intracellular and secreted apoE protein levels in the ρ0 cells and interrogated the impact of transcription factors and stress signaling pathways known to influence APOE expression. RESULTS SH-SY5Y ρ0 cells exhibited a 65-fold increase in APOE mRNA, an 8-fold increase in secreted apoE protein, and increased intracellular apoE protein. Other models of primary mitochondrial dysfunction including partial mtDNA-depletion, toxin-induced respiratory chain inhibition, and chemical-induced manipulations of the mitochondrial membrane potential similarly increased SH-SY5Y cell APOE mRNA. We explored potential mediators and found in the ρ0 cells knock-down of the C/EBPα and NFE2L2 (Nrf2) transcription factors reduced APOE mRNA. The activity of two mitogen-activated protein kinases, JNK and ERK, also strongly influenced ρ0 cell APOE mRNA levels. CONCLUSION Primary mitochondrial dysfunction either directly or indirectly activates APOE expression in a neuronal cell model by altering transcription factors and stress signaling pathways. These studies demonstrate mitochondrial biology can influence the biology of the APOE gene and apoE protein, which are implicated in AD.
Collapse
Affiliation(s)
- Alexander P Gabrielli
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ian Weidling
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Amol Ranjan
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Xiaowan Wang
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Lesya Novikova
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Subir Roy Chowdhury
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
| | - Blaise Menta
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alexandra Berkowicz
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kenneth R Peterson
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
57
|
Wen J, Bao Z, Li L, Liu Y, Wei B, Ye X, Xu H, Cui L, Li X, Shen G, Fang Y, Zeng H, Shen Z, Guo E, Jin H, Wu L. Qiangguyin inhibited fat accumulation in OVX mice through the p38 MAPK signaling pathway to achieve anti-osteoporosis effects. Biomed Pharmacother 2023; 158:114122. [PMID: 36566522 DOI: 10.1016/j.biopha.2022.114122] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a common bone disease characterized by decreased bone density and increased bone fragility due to decreased estrogen levels. Qiangguyin (QGY) is transformed from the famous traditional Chinese medicine BuShen Invigorating Blood Decoction. In this study, we used QGY to treat PMOP. We observed that QGY significantly reduced fat accumulation in the chondro-osseous junction. However, its specific mechanism of action remains unclear. To determine the specific molecular mechanism of QGY, we explored the pharmacological mechanism by which QGY reduces fat accumulation in the chondro-osseous junction through network pharmacological analysis. The active components and targets related to PMOP and QGY were screened from different databases, forming a composition-target-disease network. Next, a comprehensive analysis platform including protein-protein interaction (PPI) network, Gene Ontology (GO) enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were established. The results revealed that QGY inhibits adipogenic differentiation by activating the mitogen-activated protein kinase (MAPK) signaling pathway, thus reducing the accumulation of fat in the chondro-osseous junction. For further verification. In vitro and in vivo experiments were carried out. Our data showed that QGY significantly reversed the high expression of fatty acid binding protein 4 (FABP4) and peroxisome proliferator-activated receptor γ (PPARγ). Further, QGY prevents fat accumulation by inhibiting the expression of p38. In summary, the results of this study suggested that QGY-induced phenotypic changes are related to the activation of the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Jingyuan Wen
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhengsheng Bao
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lunxin Li
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingquan Liu
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bing Wei
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoang Ye
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huihui Xu
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Longkang Cui
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuefei Li
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Gaobo Shen
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Fang
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hanbing Zeng
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhe Shen
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Enping Guo
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Lianguo Wu
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
58
|
Zhang MZ, Dong XH, Zhang WC, Li M, Si LB, Liu YF, Li HR, Zhao PX, Liu MY, Adzavon YM, Wang XJ, Long X, Ding Y. A comparison of proliferation levels in normal skin, physiological scar and keloid tissue. J Plast Surg Hand Surg 2023; 57:122-128. [PMID: 34964674 DOI: 10.1080/2000656x.2021.2017294] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proliferation is an important characteristic of life, and many signaling pathways participate in this complicated process. The MAPK/Erk pathway is a classic pathway in cell proliferation. In this study, expression levels of key factors in the MAPK/Erk pathway were measured to assess the proliferation level among normal skin, physiological scar, and keloid tissue. Thirty patients were selected randomly from the Department of Plastic Surgery at Peking Union Medical College Hospital from January 2019 to December 2020. Histological appearance and fiber tissue content were observed by Hematoxylin and eosin staining and Masson staining. Expression levels of key factors in the MAPK/Erk pathway (ATF2, c-Jun, c-Myc, p38 and STAT1) and relative proteins (HIF-1α and PCNA) in tissues were detected by immunohistochemistry and analyzed as the percentage of positively stained cells in both the tissue epidermis and dermis. Western blot was used for quantitative analysis of the above factors. In results, keloid tissue showed a significantly higher fiber and less cell content. In the immunohistochemical result, higher expression of key factors was observed in the epidermis than in the dermal layer, and the expression of all factors was increased remarkably in keloid tissue. In western blot analysis, all factors (except STAT1) showed higher expression in keloid tissue. In our former research, keloid showed similar apoptosis level as physiological scar and normal skin. On combining our former conclusion and results in this study, an imbalance condition between the high proliferation level and normal apoptosis level may lead to the growth characteristics of keloid.
Collapse
Affiliation(s)
- Ming-Zi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Xin-Hang Dong
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Chao Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Ming Li
- Department of Orthopaedics, Qingdao Huangdao District Hospital of Traditional Chinese Medicine, Qingdao, China
| | - Lou-Bin Si
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yi-Fang Liu
- International Education College, Beijing Vocational College of Agriculture, Beijing, China
| | - Hao-Ran Li
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng-Xiang Zhao
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Meng-Yu Liu
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | | | - Xiao-Jun Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yu Ding
- Department of Information Engineering, Chaoshan Polytechnic College, Puning, China
| |
Collapse
|
59
|
Jangam A, Tirunavalli SK, Adimoolam BM, Kasireddy B, Patnaik SS, Erukkambattu J, Thota JR, Andugulapati SB, Addlagatta A. Anti-inflammatory and antioxidant activities of Gymnema Sylvestre extract rescue acute respiratory distress syndrome in rats via modulating the NF-κB/MAPK pathway. Inflammopharmacology 2023; 31:823-844. [PMID: 36662401 PMCID: PMC9864508 DOI: 10.1007/s10787-022-01133-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/30/2022] [Indexed: 01/21/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is one of the major causes of mortality in COVID-19 patients, due to limited therapeutic options. This prompted us to explore natural sources to mitigate this condition. Gymnema Sylvestre (GS) is an ancient medicinal plant known to have various therapeutic effects. This investigation examined the therapeutic effect of hydroalcoholic extract of Gymnema Sylvestre (HAEGS) against lipopolysaccharide (LPS)-induced lung injury and ARDS in in vitro and in vivo models. UHPLC-HRMS/GC-MS was employed for characterizing the HAEGS and identified several active derivatives including gymnemic acid, gymnemasaponins, gymnemoside, gymnemasin, quercetin, and long fatty acids. Gene expression by RT-qPCR and DCFDA analysis by flow cytometry revealed that several inflammatory cytokine/chemokine, cell injury markers, and reactive oxygen species (ROS) levels were highly upregulated in LPS control and were significantly reduced upon HAEGS treatment. Consistent with the in vitro studies, we found that in LPS-induced ARDS model, pre-treatment with HAEGS significantly suppressed the LPS-induced elevation of inflammatory cell infiltrations, cytokine/chemokine marker expression, ROS levels, and lung injury in a dose-dependent manner. Further mechanistic studies demonstrated that HAEGS suppressed oxidative stress by modulating the NRF2 pathway and ameliorated the ARDS through the NF-κB/MAPK signalling pathway. Additional fractionation results revealed that fraction 6 which has the exclusive composition of gymnemic acid derivatives showed better anti-inflammatory effects (inhibition of IL-6 and IL-1β) at lower concentrations compared to HAEGS. Overall, HAEGS significantly mitigated LPS-induced lung injury and ARDS by targeting the NF-κB/MAPK signalling pathway. Thus, our work unravels the protective role of HAEGS for the first time in managing ARDS.
Collapse
Affiliation(s)
- Aruna Jangam
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| | - Satya Krishna Tirunavalli
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| | - Bala Manikantha Adimoolam
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| | - Bhavana Kasireddy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India
| | - Samata Sai Patnaik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India
| | - Jayashankar Erukkambattu
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh 462020 India
| | - Jagadeshwar Reddy Thota
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India ,Department of Analytical and Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Sai Balaji Andugulapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| | - Anthony Addlagatta
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500007 India ,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| |
Collapse
|
60
|
Zhao S, Zhang D, Liu S, Huang J. The roles of NOP56 in cancer and SCA36. Pathol Oncol Res 2023; 29:1610884. [PMID: 36741964 PMCID: PMC9892063 DOI: 10.3389/pore.2023.1610884] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023]
Abstract
NOP56 is a highly conserved nucleolar protein. Amplification of the intron GGCCTG hexanucleotide repeat sequence of the NOP56 gene results in spinal cerebellar ataxia type 36 (SCA36). NOP56 contains an N-terminal domain, a coiled-coil domain, and a C-terminal domain. Nucleolar protein NOP56 is significantly abnormally expressed in a number of malignant tumors, and its mechanism is different in different tumors, but its regulatory mechanism in most tumors has not been fully explored. NOP56 promotes tumorigenesis in some cancers and inhibits tumorigenesis in others. In addition, NOP56 is associated with methylation in some tumors, suggesting that NOP56 has the potential to become a tumor-specific marker. This review focuses on the structure, function, related signaling pathways, and role of NOP56 in the progression of various malignancies, and discusses the progression of NOP56 in neurodegenerative and other diseases.
Collapse
Affiliation(s)
- Shimin Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongdong Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sicheng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China,*Correspondence: Jun Huang,
| |
Collapse
|
61
|
Zhang Q, Ren J, Wang F, Li M, Pan M, Zhang H, Qu F. Chinese herbal medicine alleviates the pathogenesis of polycystic ovary syndrome by improving oxidative stress and glucose metabolism via mitochondrial Sirtuin 3 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154556. [PMID: 36610149 DOI: 10.1016/j.phymed.2022.154556] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/29/2022] [Accepted: 11/11/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders among women, and the curative effects of its current management are not satisfactory. A formula of Chinese herbal medicine (CHM), called Bu-Shen-Tian-Jing Formula (BSTJF), has clinically shown beneficial effects in treating PCOS. PURPOSE This study aimed to investigate the mechanism underlying BSTJF for treatment of PCOS. METHODS Whole blood samples were collected from women with PCOS treated and not treated with BSTJF (n = 5 per group). Whole transcriptome sequencing of leukocytes and untargeted metabonomic analysis of the plasma were performed. Three groups of 18 female Sprague-Dawley rats were randomly selected: control, PCOS, and BSTJF. A PCOS rat model was established using testosterone propionate. The estrous cycle; glucose tolerance; ovarian morphology; serum markers of oxidative stress; and expression of Sirtuin 3 (SIRT3), phospho-p38 mitogen-activated protein kinase, phosphatidylinositol 3-kinase (PI3K), and phospho-protein kinase B in the ovary were measured. Palmitate was initially applied to KGN cells, followed by freeze-dried BSTJF powder. The glucose uptake, reactive oxygen species (ROS) production, and protein levels of SIRT3, PI3K, and glucose transporter type 4 (GLUT4) were detected in KGN cells. RESULTS The transcriptomic and metabolomic profiles showed alterations in 572 genes and 73 metabolites in women with PCOS treated with BSTJF. The enriched pathways in women with PCOS treated with BSTJF were mainly involved in inflammation, insulin resistance, glucose and lipid metabolism, and neuro and associated signaling pathways. In PCOS rat models, BSTJF improved the estrous cycle, glucose tolerance, and ovarian morphology; relieved oxidative stress; increased ovarian SIRT3 expression; inhibited p38 MAPK activation; and promoted the activation of PI3K/AKT signaling in the ovary. In the in-vitro study with KGN cells, BSTJF rescued the palmitate-induced impaired glucose uptake and SIRT3 expression, reduced mitochondrial ROS production mediated by SIRT3, and restored the impaired insulin-induced PI3K/AKT signaling pathway. CONCLUSION BSTJF effectively alleviated the pathogenesis of PCOS by improving oxidative stress and glucose metabolism via mitochondrial SIRT3 and the following insulin signaling pathway. This study innovatively revealed the action mechanism of CHM in treating PCOS.
Collapse
Affiliation(s)
- Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China; Zhejiang Hospital, Hangzhou, Zhejiang 310000, China
| | - Jun Ren
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| | - Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| | - Mingqian Li
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, China
| | - Manman Pan
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| | - Hui Zhang
- Zhejiang Vocational College of Special Education, Hangzhou, Zhejiang 310023, China
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
62
|
Yi Z, Jia Q, Lin Y, Wang Y, Cong J, Gu Z, Ling J, Cai G. Mechanism of Elian granules in the treatment of precancerous lesions of gastric cancer in rats through the MAPK signalling pathway based on network pharmacology. PHARMACEUTICAL BIOLOGY 2022; 60:87-95. [PMID: 34962453 PMCID: PMC8725869 DOI: 10.1080/13880209.2021.2017980] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/17/2021] [Accepted: 12/08/2021] [Indexed: 05/26/2023]
Abstract
CONTEXT Elian Granules have been applied in the treatment of precancerous lesions of gastric cancer (PLGC) and achieved good results. However, its exact mechanism remains unclear. OBJECTIVES To explore the mechanism of Elian granules in treating PLGC through the mitogen-activated protein kinase (MAPK) signalling pathway based on network pharmacology. MATERIALS AND METHODS Through network pharmacological methods, the targets of the active component of Elian granules against PLGC were obtained. Subsequently, Specific Pathogen Free (SPF) male Sprague Dawley (SD) rats were randomly divided into normal, model, and Elian granule groups. The N-methyl-N'-nitro-N-nitrosoguanidine comprehensive method was used to establish the PLGC rat model. The model and Elian granule groups were given normal saline and Elian granule aqueous solution (3.24 g/kg/d) intragastric administration, respectively, for 24 weeks. The pathological changes in gastric tissues were observed by hematoxylin-eosin staining. The protein expression of p-JNK and p-p38 was verified by western blotting. RESULTS 394 and 4,395 targets were identified in Elian granules and PLGC, respectively. The 190 common targets were mainly enriched in MAPK signalling pathways. The gastric mucosal epithelium was still intact, the glands were arranged regularly, and no goblet cells or apparent inflammatory cell infiltration were observed in the Elian granule group. The expression of p-JNK and p-p38 protein of the Elian granule group (0.83 ± 0.08; 1.18 ± 0.40) was significantly higher than the model group (0.27 ± 0.14; 0.63 ± 0.14) (p < 0.01; p < 0.05). DISCUSSION AND CONCLUSIONS Elian granules may play a critical role in the treatment of rat PLGC by up-regulating the expression of p-JNK and p-p38 proteins in the MAPK signalling pathway, thus providing a scientific basis for clinical application.
Collapse
Affiliation(s)
- Zhirong Yi
- The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People’s Republic of China
| | - Qingling Jia
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yili Lin
- The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People’s Republic of China
| | - Yujiao Wang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jun Cong
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhijian Gu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jianghong Ling
- The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, People’s Republic of China
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Gan Cai
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
63
|
Wang C, Li Y, Yi Y, Liu G, Guo R, Wang L, Lan T, Wang W, Chen X, Chen S, Yu SY. Hippocampal microRNA-26a-3p deficit contributes to neuroinflammation and behavioral disorders via p38 MAPK signaling pathway in rats. J Neuroinflammation 2022; 19:283. [PMID: 36434679 PMCID: PMC9694101 DOI: 10.1186/s12974-022-02645-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/15/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Neuronal injury is considered a critical risk factor in the pathogenesis of most neurological and neuropsychiatric diseases. However, the underlying molecular mechanisms and identification of potential therapeutic targets for preventing neuronal injury associated with brain function remain largely uncharacterized. Therefore, identifying neural mechanisms would put new insights into the progression of this condition and provide novel therapeutic strategies for the treatment of these diseases. METHODS Stereotactic injection of AAV virus was used to knock-down the miR-26a-3p within hippocampus of rats. Behavioral changes was detected by open field test (OFT), elevated plus maze (EPM), forced swim test (FST) and sucrose preference test (SPT). The inflammatory cytokines and related proteins were verified by real-time quantitative PCR, immunoblotting or immunofluorescence assay. Golgi staining and electron microscopy analysis was used to observe the dendritic spine, synapse and ultrastructural pathology. SB203580 (0.5 mg/kg) were administered daily to prevent p38 MAPK via an intraperitoneal (i.p.) injection. Finally, electrophysiological method was used to examine the synaptic transmission via whole-cell patch-clamp recording. RESULTS Here, we showed that miR-26a-3p deficiency within hippocampal regions leads to the activation of microglia, increased level of pro-inflammatory cytokines and behavioral disorders in rats, effects which appear to be mediated by directly targeting the p38 mitogen-activated protein kinase (MAPK)-NF-κB signaling pathway. Specifically, we found that the enhanced glia-activation may consequently result in neuronal deterioration that mainly presented as the dysregulation of structural and functional plasticity in hippocampal neurons. In contrast, preventing p38 pathway by SB203580 significantly ameliorated abnormal behavioral phenotypes and neuronal jury resulting from miR-26a-3p knock-down. CONCLUSION These results suggest that the normal expression of miR-26a-3p exerts neuroprotective effects via suppressing neural abnormality and maintaining neuroplasticity to against behavioral disorders in rats. These effects appear to involve a down-regulation of p38 MAPK-NF-κB signaling within the hippocampal region. Taken together, these findings provide evidence that miR-26a-3p can function as a critical factor in regulating neural activity and suggest that the maintaining of normal structure and function of neurons might be a potential therapeutic strategy in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Changmin Wang
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Ye Li
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Yuhang Yi
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Guiyu Liu
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Ruojing Guo
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Liyan Wang
- grid.27255.370000 0004 1761 1174Morphological Experimental Center, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Tian Lan
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Wenjing Wang
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Xiao Chen
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| | - Shihong Chen
- grid.27255.370000 0004 1761 1174Department of Endocrinology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, 250033 Shandong People’s Republic of China
| | - Shu Yan Yu
- grid.27255.370000 0004 1761 1174Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China ,Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, 250012 Shandong People’s Republic of China
| |
Collapse
|
64
|
Grave N, Scheffel TB, Cruz FF, Rockenbach L, Goettert MI, Laufer S, Morrone FB. The functional role of p38 MAPK pathway in malignant brain tumors. Front Pharmacol 2022; 13:975197. [PMID: 36299892 PMCID: PMC9589890 DOI: 10.3389/fphar.2022.975197] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Gliomas are extremely debilitating malignant brain tumors with very limited response to therapies. The initiation and progression of gliomas can be attributed to several molecular abnormalities, such as mutations in important regulatory networks. In this regard, the mitogen-activated protein kinases (MAPKs) arise as key signaling pathways involved in cell proliferation, survival, and differentiation. MAPK pathway has been altered in most glial tumors. In glioma cells, the activation of p38 MAPK contributes to tumor invasion and metastasis and is positively correlated with tumor grade, being considered a potential oncogenic factor contributing to brain tumorigenesis and chemotherapy resistance. Hence, a better understanding of glioma pathogenesis is essential to the advancement of therapies that provide extended life expectancy for glioma patients. This review aims to explore the role of the p38 MAPK pathway in the genesis and progression of malignant brain tumors.
Collapse
Affiliation(s)
- Nathália Grave
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thamiris Becker Scheffel
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Fernandes Cruz
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Liliana Rockenbach
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcia Inês Goettert
- Laboratorio de Cultura de Células, Programa de Pós-Graduação em Biotecnologia, Universidade do Vale do Taquari (Univates), Lajeado, Brazil
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Fernanda Bueno Morrone
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- *Correspondence: Fernanda Bueno Morrone,
| |
Collapse
|
65
|
Yu H, Su X, Lei T, Zhang L, Feng Z, Zhang C, Zhang M, Wang Y, Chen X, Liu J. Safety and efficacy of p38 mitogen-activated protein kinase inhibitors (MAPKIs) in COPD. Front Pharmacol 2022; 13:950035. [PMID: 36249771 PMCID: PMC9554617 DOI: 10.3389/fphar.2022.950035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Chronic inflammation is the core mechanism of the development of chronic obstructive pulmonary disease (COPD). Corticosteroid resistance in COPD limits its anti-inflammatory potency. p38 MAPKIs were suggested as an alternative to corticosteroids despite the fact that there is currently no systematic review evaluating existing evidence.Methods: This randomized controlled trials (RCT)-based systematic review with meta-analysis was conducted following the PRISMA statement. RCTs were searched and screened from 8 databases. Three types of data, including basic information of included studies, pre-defined outcome data, and quality assessment information were extracted. Pooling values and associated 95 % confidence intervals were deemed as statistically significant only when two-tailed p values were smaller than 0.05.Results: This study included 10 RCTs with a total population of 1,751 [age, mean (SD) = 64.39 (8.06)]. Safety and several efficacy indicators of lung function, inflammatory biomarkers, and quality of life were meta-analyzed. Despite the improvement of post-bronchodilator-forced vital capacity (FVC), no difference between p38 MAPKIs and placebo was found in both safety and efficacy.Conclusion: Compared with placebo, p38 MAPKIs are safe but did not show any significant effects in the COPD population. Results of this study should be regarded with caution due to the small number of included studies and heterogeneity from combining different p38 MAPKIs as a whole.Systematic Review registration: PROSPERO #CRD42022302890.
Collapse
Affiliation(s)
- Haichuan Yu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaojie Su
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ting Lei
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lu Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhouzhou Feng
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Chuchu Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Meng Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yalei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xinlong Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Jian Liu,
| |
Collapse
|
66
|
Hu H, Hu Z, Zhang Y, Wan H, Yin Z, Li L, Liang X, Zhao X, Yin L, Ye G, Zou YF, Tang H, Jia R, Chen Y, Zhou H, Song X. Myricetin inhibits pseudorabies virus infection through direct inactivation and activating host antiviral defense. Front Microbiol 2022; 13:985108. [PMID: 36187970 PMCID: PMC9520584 DOI: 10.3389/fmicb.2022.985108] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022] Open
Abstract
Myricetin, a polyhydroxyflavone compound, is one of the main ingredients of various human foods and therefore also known as dietary flavonoids. Due to the continuous emergence of resistant strains of herpesviruses, novel control measures are required. In the present study, myricetin exhibited potent antiviral activity against pseudorabies virus (PRV), a model organism of herpesvirus. The suppression rate could reach up to 96.4% at a concentration of 500 μM in cells, and the 50% inhibitory concentration (IC50) was 42.69 μM. Moreover, the inhibitory activity was not attenuated by the increased amount of infective dose, and a significant reduction of intracellular PRV virions was observed by indirect immunofluorescence. A mode of action study indicated that myricetin could directly inactivate the virus in vitro, leading to inhibition of viral adsorption, penetration and replication in cells. In addition to direct killing effect, myricetin could also activate host antiviral defense through regulation of apoptosis-related gene expressions (Bcl-2, Bcl-xl, Bax), NF-κB and MAPK signaling pathways and cytokine gene expressions (IL-1α, IL-1β, IL-6, c-Jun, STAT1, c-Fos, and c-Myc). In PRV-infected mouse model, myricetin could enhance the survival rate by 40% at 5 days post infection, and viral loads in kidney, liver, lung, spleen, and brain were significantly decreased. The pathological changes caused by PRV infection were improved by myricetin treatment. The gene expressions of inflammatory factors (MCP-1, G-CSF, IL-1α, IL-1β, and IL-6) and apoptotic factors (Bcl-xl, Bcl-2, and Bax) were regulated by myricetin in PRV-infected mice. The present findings suggest that myricetin can effectively inhibit PRV infection and become a candidate for development of new anti-herpesvirus drugs.
Collapse
Affiliation(s)
- Huaiyue Hu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhiqiang Hu
- Shandong New Hope Liuhe Agriculture and Animal Husbandry Technology Co., Ltd., Dezhou, China
| | - Yingying Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hongping Wan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinghong Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuan-Feng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yaqin Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hao Zhou
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
67
|
Tizaoui K, Shin JI, Jeong GH, Yang JW, Park S, Kim JH, Hwang SY, Park SJ, Koyanagi A, Smith L. Genetic Polymorphism of PTPN22 in Autoimmune Diseases: A Comprehensive Review. Medicina (B Aires) 2022; 58:medicina58081034. [PMID: 36013501 PMCID: PMC9415475 DOI: 10.3390/medicina58081034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 12/04/2022] Open
Abstract
It is known that the etiology and clinical outcomes of autoimmune diseases are associated with a combination of genetic and environmental factors. In the case of the genetic factor, the SNPs of the PTPN22 gene have shown strong associations with several diseases. The recent exploding numbers of genetic studies have made it possible to find these associations rapidly, and a variety of autoimmune diseases were found to be associated with PTPN22 polymorphisms. Proteins encoded by PTPN22 play a key role in the adaptative and immune systems by regulating both T and B cells. Gene variants, particularly SNPs, have been shown to significantly disrupt several immune functions. In this review, we summarize the mechanism of how PTPN22 and its genetic variants are involved in the pathophysiology of autoimmune diseases. In addition, we sum up the findings of studies reporting the genetic association of PTPN22 with different types of diseases, including type 1 diabetes mellitus, systemic lupus erythematosus, juvenile idiopathic arthritis, and several other diseases. By understanding these findings comprehensively, we can explain the complex etiology of autoimmunity and help to determine the criteria of disease diagnosis and prognosis, as well as medication developments.
Collapse
Affiliation(s)
- Kalthoum Tizaoui
- Department of Basic Sciences, Division of Histology and Immunology, Faculty of Medicine Tunis, Tunis El Manar University, Tunis 2092, Tunisia;
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Gwang Hun Jeong
- College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea;
| | - Seoyeon Park
- Yonsei University College of Medicine, Seoul 06273, Korea; (S.P.); (S.Y.H.)
| | - Ji Hong Kim
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea;
- Correspondence: ; Tel.: +82-2-2019-3352; Fax: +82-2-3461-9473
| | - Soo Young Hwang
- Yonsei University College of Medicine, Seoul 06273, Korea; (S.P.); (S.Y.H.)
| | - Se Jin Park
- Department of Pediatrics, Eulji University School of Medicine, Daejeon 35233, Korea;
| | - Ai Koyanagi
- Research and Development Unit, Parc Sanitari Sant Joan de Déu, CIBERSAM, Dr. Antoni Pujadas, 42, Sant Boi de Llobregat, 08830 Barcelona, Spain;
- ICREA, Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Lee Smith
- Centre for Health Performance and Wellbeing, Anglia Ruskin University, Cambridge CB1 1PT, UK;
| |
Collapse
|
68
|
Zileuton Alleviates Radiation-Induced Cutaneous Ulcers via Inhibition of Senescence-Associated Secretory Phenotype in Rodents. Int J Mol Sci 2022; 23:ijms23158390. [PMID: 35955523 PMCID: PMC9369445 DOI: 10.3390/ijms23158390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation-induced cutaneous ulcers are a challenging medical problem for patients receiving radiation therapy. The inhibition of cell senescence has been suggested as a prospective strategy to prevent radiation ulcers. However, there is no effective treatment for senescent cells in radiation ulcers. In this study, we investigated whether zileuton alleviated radiation-induced cutaneous ulcer by focusing on cell senescence. We demonstrate increased cell senescence and senescence-associated secretory phenotype (SASP) in irradiated dermal fibroblasts and skin tissue. The SASP secreted from senescent cells induces senescence in adjacent cells. In addition, 5-lipoxygenase (5-LO) expression increased in irradiated dermal fibroblasts and skin tissue, and SASP and cell senescence were regulated by 5-LO through p38 phosphorylation. Finally, the inhibition of 5-LO following treatment with zileuton inhibited SASP and mitigated radiation ulcers in animal models. Our results demonstrate that inhibition of SASP from senescent cells by zileuton can effectively mitigate radiation-induced cutaneous ulcers, indicating that inhibition of 5-LO might be a viable strategy for patients with this condition.
Collapse
|
69
|
Wang J, Chen S, Pan C, Li G, Tang Z. Application of Small Molecules in the Central Nervous System Direct Neuronal Reprogramming. Front Bioeng Biotechnol 2022; 10:799152. [PMID: 35875485 PMCID: PMC9301571 DOI: 10.3389/fbioe.2022.799152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of regenerative capacity of neurons leads to poor prognoses for some neurological disorders. The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases. In this review, the mechanisms of action of different small molecules, the approaches to screening small molecule cocktails, and the methods employed to detect their reprogramming efficiency are discussed, and the studies, focusing on neuronal reprogramming using small molecules in neurological disease models, are collected. Future research efforts are needed to investigate the in vivo mechanisms of small molecule-mediated neuronal reprogramming under pathophysiological states, optimize screening cocktails and dosing regimens, and identify safe and effective delivery routes to promote neural regeneration in different neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Gaigai Li
- *Correspondence: Gaigai Li, ; Zhouping Tang,
| | | |
Collapse
|
70
|
Alpha1B-adreneroceptor is involved in norepinephrine-induced pulmonary artery smooth muscle cell proliferation via p38 signaling. Eur J Pharmacol 2022; 931:175159. [DOI: 10.1016/j.ejphar.2022.175159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
|
71
|
He S, Ning Y, Ma F, Liu D, Jiang S, Deng S. IL-23 Inhibits Trophoblast Proliferation, Migration, and EMT via Activating p38 MAPK Signaling Pathway to Promote Recurrent Spontaneous Abortion. J Microbiol Biotechnol 2022; 32:792-799. [PMID: 35637168 PMCID: PMC9628909 DOI: 10.4014/jmb.2112.12056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/15/2022]
Abstract
As a vital problem in reproductive health, recurrent spontaneous abortion (RSA) affects about 1% of women. We performed this study with an aim to explore the molecular mechanism of interleukin-23 (IL-23) and find optimal or effective methods to improve RSA. First, ELISA was applied to evaluate the expressions of IL-23 and its receptor in HTR-8/SVneo cells after IL-23 treatment. CCK-8, TUNEL, wound healing and transwell assays were employed to assess the proliferation, apoptosis, migration and invasion of HTR-8/SVneo cells, respectively. Additionally, the expressions of apoptosis-, migration-, epithelial-mesenchymal transition- (EMT-) and p38 MAPK signaling pathway-related proteins were measured by western blotting. To further investigate the relationship between IL-23 and p38 MAPK signaling pathway, HTR-8/SVneo cells were treated for 1 h with p38 MAPK inhibitor SB239063, followed by a series of cellular experiments on proliferation, apoptosis, migration and invasion, as aforementioned. The results showed that IL-23 and its receptors were greatly elevated in IL-23-treated HTR-8/SVneo cells. Additionally, IL-23 demonstrated suppressive effects on the proliferation, apoptosis, migration, invasion and EMT of IL-23-treated HTR-8/SVneo cells. More importantly, the molecular mechanism of IL-23 was revealed in this study; that is to say, IL-23 inhibited the proliferation, apoptosis, migration, invasion and EMT of IL-23-treated HTR-8/SVneo cells via activating p38 MAPK signaling pathway. In conclusion, IL-23 inhibits trophoblast proliferation, migration, and EMT via activating p38 MAPK signaling pathway, suggesting that IL-23 might be a novel target for the improvement of RSA.
Collapse
Affiliation(s)
- Shan He
- Department of Pharmacy, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Yan Ning
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China,Corresponding author Phone: +0755-82889999 E-mail:
| | - Fei Ma
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Dayan Liu
- Department of Genesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Shaoyan Jiang
- Department of Pharmacy, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Shaojie Deng
- Department of Pharmacy, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| |
Collapse
|
72
|
Hashimoto T, Kondo N, Makino A, Kiyono Y, Temma T. Radiobrominated probe targeting activated p38α in inflammatory diseases. Ann Nucl Med 2022; 36:845-852. [DOI: 10.1007/s12149-022-01764-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022]
|
73
|
Singlet Oxygen, Photodynamic Therapy, and Mechanisms of Cancer Cell Death. JOURNAL OF ONCOLOGY 2022; 2022:7211485. [PMID: 35794980 PMCID: PMC9252714 DOI: 10.1155/2022/7211485] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 01/06/2023]
Abstract
Photodynamic therapy (PDT) can be developed into an important arsenal against cancer; it is a minimally invasive therapy, which is used in the treatment or/and palliation of a variety of cancers and benign diseases. The removal of cancerous tissue is achieved with the use of photosensitizer and a light source, which excites the photosensitizer. This excitation causes the photosensitizer to generate singlet oxygen and other reactive oxygen species. PDT has been used in several types of cancers including nonmelanoma skin cancer, bladder cancer, esophageal cancer, head and neck cancer, and non-small cell lung cancer (NSCLC). Although it is routinely used in nonmelanoma skin cancer, it has not been widely adopted in other solid cancers due to a lack of clinical data showing the superiority of PDT over other forms of treatment. Singlet oxygen used in PDT can alter the activity of the catalase, which induces immunomodulation through HOCl signaling. The singlet oxygen can induce apoptosis through both the extrinsic and intrinsic pathways. The extrinsic pathway of apoptosis starts with the activation of the Fas receptor by singlet oxygen that leads to activation of the caspase-7 and caspase-3. In the case of the intrinsic pathway, disruption caused by singlet oxygen in the mitochondria membrane leads to the release of cytochrome c, which binds with APAF-1 and procaspase-9, forming a complex, which activates caspase-3. Mechanisms of PDT action can vary according to organelles affected. In the plasma membrane, membrane disruption is caused by the oxidative stress leading to the intake of calcium ions, which causes swelling and rupture of cells due to excess intake of water, whereas disruption of lysosome causes the release of the cathepsins B and D, which cleave Bid into tBid, which changes the mitochondrial outer membrane permeability (MOMP). Oxidative stress causes misfolding of protein in the endoplasmic reticulum. When misfolding exceeds the threshold, it triggers unfolding protein response (UPR), which leads to activation of caspase-9 and caspase-3. Finally, the activation of p38 MAPK works as an alternative pathway for the induction of MOMP.
Collapse
|
74
|
George S, Jean-Baptiste W, Yusuf Ali A, Inyang B, Koshy FS, George K, Poudel P, Chalasani R, Goonathilake MR, Waqar S, Mohammed L. The Role of Type 2 Diabetes in Pancreatic Cancer. Cureus 2022; 14:e26288. [PMID: 35898377 PMCID: PMC9308974 DOI: 10.7759/cureus.26288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/24/2022] [Indexed: 11/05/2022] Open
Abstract
The incidence of type 2 diabetes mellitus (T2DM) and its potential complications, such as cancers, are increasing worldwide at an astounding rate. There are many factors such as obesity, diabetes, alcohol consumption, and the adoption of sedentary lifestyles that are driving pancreatic cancer (PC) to become one of the leading causes of cancer mortality in the United States. PC is notorious for its generic symptoms and late-stage presentation with rapid metastasis. The connection between T2DM and the risk of PC development is multifaceted and complex. Some of the proposed theories reveal that chronic inflammation, insulin resistance, hyperinsulinemia, hyperglycemia, and abnormalities in the insulin and insulin-like growth factor axis (IGF) contribute to the disease association between these two conditions. This literature review aims to highlight relevant studies and explore the molecular mechanisms involved in the etiology of diabetes and its impact on PC development, as well as the role of anti-diabetic agents on PC. Despite extensive studies, the exact interaction between T2DM and PC remains obscure and will need further investigation. According to current knowledge, there is a substantial link between diabetes, obesity, and dietary patterns in the development and progression of PC. Consequently, focusing our efforts on preventive measures by reducing modifiable risk factors remains the most effective strategy to reduce the risk of PC at this time. Antidiabetic drugs can have various effects on the occurrence and prognosis of PC with metformin offering a clear benefit of inhibiting PC and insulin increasing the risk of PC. The development of future novel therapies will require a deeper knowledge of the triggering mechanisms and interplay between these two disease states.
Collapse
|
75
|
Abstract
SignificanceTo ensure their survival, cells arrest the cell division cycle when they are exposed to environmental stress. The duration of this arrest is dependent upon the time it takes a cell to adapt to a particular environment. How cells adjust the amount of time they remain arrested is not known. This study investigates the role of the phosphatase calcineurin in controlling cell cycle arrest duration in yeast. We show that calcineurin lengthens arrest by prolonging Hog1-dependent activation of the poorly characterized cyclin-dependent kinase inhibitor Cip1. Cip1 only impacts cell cycle arrest in response to stressors that robustly activate calcineurin, suggesting that Cip1 is a context-specific regulator that differentially adjusts the length of arrest depending on the particular stressor.
Collapse
|
76
|
Kang J, Zhou Y, Zhu C, Ren T, Zhang Y, Xiao L, Fang B. Ginsenoside Rg1 Mitigates Porcine Intestinal Tight Junction Disruptions Induced by LPS through the p38 MAPK/NLRP3 Inflammasome Pathway. TOXICS 2022; 10:toxics10060285. [PMID: 35736894 PMCID: PMC9228030 DOI: 10.3390/toxics10060285] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
Abstract
Inflammation leads to porcine tight junction disruption of small intestinal epithelial cells, resulting in intestinal dysfunction. Herein, we established lipopolysaccharide (LPS)-induced in-vivo and in-vitro inflammatory models. The results revealed that LPS induced tight junction disruption in IPEC-J2 cells by downregulating tight-junction-related protein zonula occludens-1 (ZO-1), occludin and claudin-1 expression, while ginsenoside Rg1 rescued such inhibition and abrogated the upregulated expression of phosphorylation p38 MAPK. The p38 MAPK inhibitor (SB203580) showed a similar effect with Rg1 and attenuated the LPS-induced inhibition of ZO-1, occludin and claudin-1 expression, which is consistent with the reduced expression of NLRP3 inflammasome and IL-1β. Furthermore, the specific inhibitors of NLRP3 and IL-1β result in increased expression of tight-junction-related protein, demonstrating that p38 MAPK signaling was associated with Rg1 suppression of tight junction disruption. Besides, LPS treatment decreased the expression of ZO-1, occludin and claudin-1 through p38 MAPK signaling, and caused abnormal morphological changes in murine ileum. Meanwhile, Rg1 attenuated the decreased expression of ZO-1, occludin and claudin-1 and partially alleviated LPS-induced morphological changes in murine ileum. In summary, these findings characterized a novel mechanism by which Rg1 alleviates LPS-induced intestinal tight junction disruption by inhibiting the p38 MAPK-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Jian Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Yanhong Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Chunyang Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Tian Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
- Correspondence: (L.X.); (B.F.)
| | - Binghu Fang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
- Correspondence: (L.X.); (B.F.)
| |
Collapse
|
77
|
Dai C, Zhu J, Huang H. 混合谱系激酶3在心血管疾病中的研究进展. CHINESE SCIENCE BULLETIN-CHINESE 2022. [DOI: 10.1360/tb-2022-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
78
|
Liu C, Jiang S, Xie H, Jia H, Li R, Zhang K, Wang N, Lin P, Yu X. Long non-coding RNA AC245100.4 contributes to prostate cancer migration via regulating PAR2 and activating p38-MAPK pathway. Med Oncol 2022; 39:94. [DOI: 10.1007/s12032-022-01689-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/21/2022] [Indexed: 12/25/2022]
|
79
|
Chen Q, Wei Y, Zhao Y, Xie X, Kuang N, Wei Y, Yu M, Hu T. Intervening Effects and Molecular Mechanism of Quercitrin on PCV2-Induced Histone Acetylation, Oxidative Stress and Inflammatory Response in 3D4/2 Cells. Antioxidants (Basel) 2022; 11:antiox11050941. [PMID: 35624806 PMCID: PMC9137775 DOI: 10.3390/antiox11050941] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Porcine circovirus type 2 (PCV2) is the main pathogen causing porcine circovirus-associated diseases (PCVD/PCVADs), and infection of the host induces immunosuppression. Since quercitrin (QUE) has anti-inflammatory and antiviral activity, it is worth exploiting in animal diseases. In this study, the interventional effects and the molecular mechanism of QUE on PCV2-induced oxidative stress and inflammatory responses in 3D4/2 cells and the modulation of histone acetylation modifications were investigated. The ROS production was measured by DCFH-DA fluorescent probes. HAT and HDAC enzyme activity were determined by ELISA. Histone acetylation, oxidative stress and inflammation-related gene expression levels were measured by q-PCR. Histone H3 and H4 (AcH3 and AcH4) acetylation, oxidative stress and inflammation-related protein expression levels were measured by Western blot. The results showed that QUE treatment at different concentrations on PCV2-infected 3D4/2 cells was able to attenuate the production of ROS. Moreover, QUE treatment could also intervene in oxidative stress and decrease the enzyme activity of HAT and the mRNA expression level of HAT1, while it increased the enzyme activity of HDAC and HDAC1 mRNA expression levels and downregulated histone H3 and H4 (AcH3 and AcH4) acetylation modification levels. In addition, QUE treatment even downregulated the mRNA expression levels of IL-6, IL-8, IκB, AKT and p38, but upregulated the mRNA expression levels of IL-10, SOD, GPx1, p65, Keap1, Nrf2, HO-1 and NQO1. As to protein expression, QUE treatment downregulated the levels of iNOS, p-p65 and IL-8 as well as the phosphorylation expression of IκB and p38, while it upregulated the levels of HO-1 and NQO1. It was shown that QUE at 25, 50 or 100 μmol/L regulated p38MAPK and PI3K/AKT signaling pathways by downregulating cellular histone acetylation modification levels while inhibiting the NF-κB inflammatory signaling pathway and activating the Nrf2/HO-1 antioxidant signaling pathway, thus regulating the production of inflammatory and antioxidant factors and exerting both anti-inflammatory and antioxidant effects.
Collapse
|
80
|
Plin5, a New Target in Diabetic Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2122856. [PMID: 35509833 PMCID: PMC9060988 DOI: 10.1155/2022/2122856] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
Abstract
Abnormal lipid accumulation is commonly observed in diabetic cardiomyopathy (DC), which can create a lipotoxic microenvironment and damage cardiomyocytes. Lipid toxicity is an important pathogenic factor due to abnormal lipid accumulation in DC. As a lipid droplet (LD) decomposition barrier, Plin5 can protect LDs from lipase decomposition and regulate lipid metabolism, which is involved in the occurrence and development of cardiovascular diseases. In recent years, studies have shown that Plin5 expression is involved in the pathogenesis of DC lipid toxicity, such as oxidative stress, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and insulin resistance (IR) and has become a key target of DC research. Therefore, understanding the relationship between Plin5 and DC progression as well as the mechanism of this process is crucial for developing new therapeutic approaches and exploring new therapeutic targets. This review is aimed at exploring the latest findings and roles of Plin5 in lipid metabolism and DC-related pathogenesis, to explore possible clinical intervention approaches.
Collapse
|
81
|
Long Noncoding RNA IGFBP7-AS1 Promotes Odontogenesis of Stem Cells from Human Exfoliated Deciduous Teeth via the p38 MAPK Pathway. Stem Cells Int 2022; 2022:9227248. [PMID: 35469296 PMCID: PMC9034958 DOI: 10.1155/2022/9227248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/21/2022] [Indexed: 12/16/2022] Open
Abstract
Stem cells from human exfoliated deciduous teeth (SHED) are attractive seed cells for dental tissue engineering. Epigenetics refers to heritable changes in gene expression patterns that do not alter DNA sequences. Long noncoding RNAs (lncRNAs) are one of the main methods of epigenetic regulation and participate in cell differentiation; however, little is known regarding the role of lncRNAs during SHED odontogenic differentiation. In this study, RNA sequencing (RNA-seq) was used to obtain the expression profile of lncRNAs and mRNAs during the odontogenic differentiation of SHED. The effect of IGFBP7-AS1 on odontogenic differentiation of SHED was assessed by alkaline phosphatase (ALP) staining, alizarin red S (ARS) staining, quantitative reverse transcription PCR (qRT-PCR), Western blot, and in vivo. The level of p38 and p-p38 protein expression was examined by Western blot, and the result was verified by adding the p38 inhibitor, SB203580. The expression profiles of lncRNAs and mRNAs were identified by RNA-seq analysis, which help us to further understand the mechanism in odontogenesis epigenetically. IGFBP7-AS1 expression was increased during odontogenic differentiation on days 7 and 14. The ALP staining, ARS staining, and expression of odontogenic markers were upregulated by overexpressing IGFBP7-AS1 in vitro, whereas the expression of osteogenesis markers was not significantly changed on mRNA level. The effect of IGFBP7-AS1 was also verified in vivo. IGFBP7-AS1 could further positively regulate odontogenic differentiation through the p38 MAPK pathway. This may provide novel targets for dental tissue engineering.
Collapse
|
82
|
Yang L, An L, Wang Y, Li J. Protective effect of isopsoralen on UVB-induced injury in HaCaT cells via the ER and p38MAPK signaling pathways. J Food Biochem 2022; 46:e14163. [PMID: 35415935 DOI: 10.1111/jfbc.14163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
This study investigated the protective effect of isopsoralen on UVB-induced damage in HaCaT cells and its molecular mechanism. The cytotoxicity of isopsoralen and its effects on the viability of HaCaT cells were examined using the MTT assay. The effects of UVB irradiation and isopsoralen on the intracellular glutathione (GSH-PX), superoxide dismutase (SOD), malondialdehyde (MDA), and reactive oxygen species (ROS) content were examined using commercially available assay kits. Further, the effects of UVB irradiation and isopsoralen on the levels of the inflammatory cytokines TNF-α, IL-6, and IL-1α were examined using enzyme-linked immunosorbent assay. Finally, we examined the effect of adding the estrogen receptor (ER) antagonist ICI182780,780 and the p38MAPK antagonist SB203580 on the changes in inflammatory cytokines induced by isopsoralen treatment and UVB irradiation. Isopsoralen pretreatment markedly inhibited UVB-induced reduction in the viability and proliferation of HaCaT cells. Isopsoralen also reduced UVB-induced increase in the expression of the inflammatory cytokines and the level of free radicals (ROS and MDA), and reversed the UVB-induced suppression of antioxidant activity. Additionally, inhibition of ER and p38MAPK via the addition of their respective antagonists reversed the observed anti-inflammatory effects of Isopsoralen. Isopsoralen can efficiently provide protection against UVB-induced damage in HaCaT cells brought about via oxidation and inflammatory reactions, and the underlying mechanisms involve the ER and p38MAPK pathways. Therefore, Isopsoralen could be used in therapeutic solutions for UVB-induced skin conditions. PRACTICAL APPLICATIONS: Isopsoralen shows antioxidant and anti-inflammatory effects. As natural, healthy, and effective additives, isopsoralen has been widely used in cosmetics and botanical medicine products. The results of this study reveal the molecular mechanisms underlying isopsoralen effects, showing that isopsoralen reverses the effects of UVB irradiation regulating ER and p38MAPK signaling pathways. Consequently, isopsoralen regulates the expression of ER and p38MAPK signaling pathways, thereby reducing the activation of antioxidant and anti-inflammatory activity. These findings suggest that isopsoralen can be used as the base ingredient for antiphotoaging cosmetics and botanical medicine products. This study provides both theoretical and experimental background for isopsoralen deep processing and utilization.
Collapse
Affiliation(s)
- Liu Yang
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, China
| | - Lifeng An
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, China
| | - Yeqiu Wang
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, China
| | - Jianmin Li
- Hospital of the First Auxiliary, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
83
|
Identification and characterization of inhibitory nanobody against p38δ. Biochem Biophys Res Commun 2022; 600:60-66. [DOI: 10.1016/j.bbrc.2022.01.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/21/2022] [Indexed: 11/21/2022]
|
84
|
Deng C, Zhang L, Ma X, Cai S, Jia Y, Zhao L. RFTN1 facilitates gastric cancer progression by modulating AKT/p38 signaling pathways. Pathol Res Pract 2022; 234:153902. [DOI: 10.1016/j.prp.2022.153902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/06/2022] [Accepted: 04/15/2022] [Indexed: 10/18/2022]
|
85
|
Ozturk S. Molecular determinants of the meiotic arrests in mammalian oocytes at different stages of maturation. Cell Cycle 2022; 21:547-571. [PMID: 35072590 PMCID: PMC8942507 DOI: 10.1080/15384101.2022.2026704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/21/2021] [Accepted: 01/03/2022] [Indexed: 01/26/2023] Open
Abstract
Mammalian oocytes undergo two rounds of developmental arrest during maturation: at the diplotene of the first meiotic prophase and metaphase of the second meiosis. These arrests are strictly regulated by follicular cells temporally producing the secondary messengers, cAMP and cGMP, and other factors to regulate maturation promoting factor (composed of cyclin B1 and cyclin-dependent kinase 1) levels in the oocytes. Out of these normally appearing developmental arrests, permanent arrests may occur in the oocytes at germinal vesicle (GV), metaphase I (MI), or metaphase II (MII) stage. This issue may arise from absence or altered expression of the oocyte-related genes playing key roles in nuclear and cytoplasmic maturation. Additionally, the assisted reproductive technology (ART) applications such as ovarian stimulation and in vitro culture conditions both of which harbor various types of chemical agents may contribute to forming the permanent arrests. In this review, the molecular determinants of developmental and permanent arrests occurring in the mammalian oocytes are comprehensively evaluated in the light of current knowledge. As number of permanently arrested oocytes at different stages is increasing in ART centers, potential approaches for inducing permanent arrests to obtain competent oocytes are discussed.
Collapse
Affiliation(s)
- Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
86
|
Xiang J, Yang Z, Zhou Q. Lidocaine relieves murine allergic rhinitis by regulating the NF-κB and p38 MAPK pathways. Exp Ther Med 2022; 23:193. [PMID: 35126696 PMCID: PMC8794549 DOI: 10.3892/etm.2022.11116] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
Allergic rhinitis (AR) is one of the most common chronic inflammatory diseases and its main feature is nasal mucositis. It has been recently revealed that lidocaine demonstrates optimal effects in the treatment of various diseases. However, a limited number of studies have examined the association between lidocaine and AR. In the present study, the AR mouse model was established to explore the effects of lidocaine in AR and to further analyze its molecular mechanism. Subsequently, different concentrations of lidocaine were provided to the animals by intranasal administration and a series of indices were assessed. The data indicated that the frequencies of mouse sneezing and nose rubbing were suppressed following an increase in lidocaine concentration. Subsequently, the number of inflammatory cells was measured. Wright's-Giemsa staining results indicated that lidocaine significantly decreased the numbers of leukocytes, eosinophils, neutrophils and lymphocytes in the nasal lavage fluid (NLF) of AR mice. In addition, the expression levels of ovalbumin (OVA)-specific immunoglobulin E (IgE), leukotriene C4 (LTC4) and certain inflammatory factors were assessed by ELISA. Lidocaine reduced OVA-specific IgE and LTC4 expression in NLF and plasma derived from AR mice. It also decreased the expression levels of IL-4, IL-5, IL-13, IL-17 and TNF-α. Lidocaine caused upregulation of IFN-γ and IL-2 expression levels. Subsequently, western blot analysis indicated that lidocaine suppressed phosphorylated (p)-p38 and p-p65 expression levels in AR mice. Collectively, the results indicated that the NF-κB and p38 MAPK signaling pathways were involved in the lidocaine-mediated relief of AR in mice. In order to further verify the association between the NF-κB and p38 MAPK signaling pathways and AR in mice, the effects of the NF-κB inhibitor IMD-0354 and the p38 MAPK inhibitor SB 203580 were assessed on AR mice. The results indicated that these two compounds exhibited similar inhibitory effects on AR mice as those noted with the use of lidocaine. These findings suggested that lidocaine represented a novel therapeutic agent for AR.
Collapse
Affiliation(s)
- Jing Xiang
- Department of Anesthesiology, Wuhan Jinyintan Hospital, Wuhan, Hubei 430000, P.R. China
| | - Zhen Yang
- Department of Anesthesiology, Wuhan Jinyintan Hospital, Wuhan, Hubei 430000, P.R. China
| | - Qiang Zhou
- Department of Anesthesiology, Wuhan Jinyintan Hospital, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
87
|
Liu K, Liu E, Lin L, Hu Y, Yuan Y, Xiao W. L-Theanine mediates the p38MAPK signaling pathway to alleviate heat-induced oxidative stress and inflammation in mice. Food Funct 2022; 13:2120-2130. [PMID: 35112126 DOI: 10.1039/d1fo03077a] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
L-Theanine, an active ingredient in the tea plant (Camellia sinensis) associated with calming, is widely used as a functional ingredient and dietary supplement. In this study, a heat stress mouse model was used to evaluate the anti-heat stress effect of L-theanine and its possible mechanism of action. Mice subjected to heat stress (40 °C) that were administered L-theanine at various doses (100, 200, and 400 mg kg-1 d-1) had reduced oxidative stress and inflammatory factors when L-theanine was administered both long-term and as a preventative treatment. Our L-theanine intervention countered the reduction in growth and feed intake of mice under heat stress and reversed liver and jejunum tissue damage. Moreover, L-theanine countered the increase in inflammatory factors TNF-α, IL-6, and IL-1β and antioxidant enzymes SOD and CAT; it also counteracted GSH-Px inactivation, the upregulation of AST and ALT enzyme activity, and MDA production. The mechanism of action may involve mediation of the P38 signaling pathway, inhibition of MK2 overexpression, and downregulation of p-P65/P65 caused by the overexpression of downstream HSP27. This would inhibit the heat stress-induced imbalance in oxidative stress and inflammatory responses.
Collapse
Affiliation(s)
- Kehong Liu
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Enshuo Liu
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Ling Lin
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Yuan Hu
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Yong Yuan
- Hunan Tea Group Co., Ltd, Changsha 410128, China
| | - Wenjun Xiao
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
- Key Lab of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China
| |
Collapse
|
88
|
Wang Y, Niu Y, Lin F, Su P, Chen L, Liu D, Sun Y. X-ray Irradiation Improves Neurological Function Recovery of Injured Spinal Cord by Inhibiting Inflammation and Glial Scar Formation. J Mol Neurosci 2022; 72:1008-1017. [DOI: 10.1007/s12031-022-01975-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
|
89
|
Lee J, Levin DE. Differential metabolism of arsenicals regulates Fps1-mediated arsenite transport. J Cell Biol 2022; 221:212996. [PMID: 35139143 PMCID: PMC8932518 DOI: 10.1083/jcb.202109034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/18/2021] [Accepted: 12/27/2021] [Indexed: 01/21/2023] Open
Abstract
Arsenic is an environmental toxin that exists mainly as pentavalent arsenate and trivalent arsenite. Both forms activate the yeast SAPK Hog1 but with different consequences. We describe a mechanism by which cells distinguish between these arsenicals through one-step metabolism to differentially regulate the bidirectional glycerol channel Fps1, an adventitious port for arsenite. Cells exposed to arsenate reduce it to thiol-reactive arsenite, which modifies a set of cysteine residues in target proteins, whereas cells exposed to arsenite metabolize it to methylarsenite, which modifies an additional set of cysteine residues. Hog1 becomes arsenylated, which prevents it from closing Fps1. However, this block is overcome in cells exposed to arsenite through methylarsenylation of Acr3, an arsenite efflux pump that we found also regulates Fps1 directly. This adaptation allows cells to restrict arsenite entry through Fps1 and also allows its exit when produced from arsenate exposure. These results have broad implications for understanding how SAPKs activated by diverse stressors can drive stress-specific outputs.
Collapse
Affiliation(s)
- Jongmin Lee
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, MA
| | - David E Levin
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, MA.,Department of Microbiology, Boston University School of Medicine, Boston, MA
| |
Collapse
|
90
|
Shanta Isl M, Arooj M, Ali I, Dong Su X, Young Yang S, Ho Kim Y, Koh YS. Anti-Inflammatory Activity of (2E)-7-hydroxy-3,7-dimethyl-2- octenyl-β-D-Glucopyranoside, Isolated from Sanguisorba officinalis L. in Toll-Like Receptor 9-Stimulated Immune Cells. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.87.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
91
|
Gutierrez DA, Contreras L, Villanueva PJ, Borrego EA, Morán-Santibañez K, Hess JD, DeJesus R, Larragoity M, Betancourt AP, Mohl JE, Robles-Escajeda E, Begum K, Roy S, Kirken RA, Varela-Ramirez A, Aguilera RJ. Identification of a Potent Cytotoxic Pyrazole with Anti-Breast Cancer Activity That Alters Multiple Pathways. Cells 2022; 11:254. [PMID: 35053370 PMCID: PMC8773755 DOI: 10.3390/cells11020254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 01/07/2022] [Indexed: 11/26/2022] Open
Abstract
In this study, we identified a novel pyrazole-based derivative (P3C) that displayed potent cytotoxicity against 27 human cancer cell lines derived from different tissue origins with 50% cytotoxic concentrations (CC50) in the low micromolar and nanomolar range, particularly in two triple-negative breast cancer (TNBC) cell lines (from 0.25 to 0.49 µM). In vitro assays revealed that P3C induces reactive oxygen species (ROS) accumulation leading to mitochondrial depolarization and caspase-3/7 and -8 activation, suggesting the participation of both the intrinsic and extrinsic apoptotic pathways. P3C caused microtubule disruption, phosphatidylserine externalization, PARP cleavage, DNA fragmentation, and cell cycle arrest on TNBC cells. In addition, P3C triggered dephosphorylation of CREB, p38, ERK, STAT3, and Fyn, and hyperphosphorylation of JNK and NF-kB in TNBC cells, indicating the inactivation of both p38MAPK/STAT3 and ERK1/2/CREB signaling pathways. In support of our in vitro assays, transcriptome analyses of two distinct TNBC cell lines (MDA-MB-231 and MDA-MB-468 cells) treated with P3C revealed 28 genes similarly affected by the treatment implicated in apoptosis, oxidative stress, protein kinase modulation, and microtubule stability.
Collapse
Affiliation(s)
- Denisse A. Gutierrez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Lisett Contreras
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Paulina J. Villanueva
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Edgar A. Borrego
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Karla Morán-Santibañez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Jessica D. Hess
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Rebecca DeJesus
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Manuel Larragoity
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Ana P. Betancourt
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Jonathon E. Mohl
- Department of Bioinformatics, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA;
| | - Elisa Robles-Escajeda
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Khodeza Begum
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Sourav Roy
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Robert A. Kirken
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Armando Varela-Ramirez
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| | - Renato J. Aguilera
- Cellular Characterization and Biorepository Core Facility, Border Biomedical Research Center, Department of Biological Sciences, College of Science, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968-0519, USA; (D.A.G.); (L.C.); (P.J.V.); (E.A.B.); (K.M.-S.); (J.D.H.); (R.D.); (M.L.); (A.P.B.); (E.R.-E.); (K.B.); (S.R.); (R.A.K.); (A.V.-R.)
| |
Collapse
|
92
|
Lim JS, Oh J, Yun HS, Lee JS, Hahn D, Kim JS. Anti-neuroinflammatory activity of 6,7-dihydroxy-2,4-dimethoxy phenanthrene isolated from Dioscorea batatas Decne partly through suppressing the p38 MAPK/NF-κB pathway in BV2 microglial cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114633. [PMID: 34520827 DOI: 10.1016/j.jep.2021.114633] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizome of Dioscorea batatas Decne (called Chinses yam) widely distributed in East Asian countries including China, Japan, Korea and Taiwan has long been used in oriental folk medicine owing to its tonic, antitussive, expectorant and anti-ulcerative effects. It has been reported to have anti-inflammatory, antioxidative, cholesterol-lowering, anticholinesterase, growth hormone-releasing, antifungal and immune cell-stimulating activities. AIM OF THE STUDY Neuroinflammation caused by activated microglia contributes to neuronal dysfunction and neurodegeneration. In the present study, the anti-neuroinflammatory activity of 6,7-dihydroxy-2,4-dimethoxy phenanthrene (DHDMP), a phenanthrene compound isolated from Dioscorea batatas Decne, was examined in microglial and neuronal cells. MATERIALS AND METHODS A natural phenanthrene compound, DHDMP, was isolated from the peel of Dioscorea batatas Decne. The anti-neuroinflammatory capability of the compound was examined using the co-culture system of BV2 murine microglial and HT22 murine neuronal cell lines. The expression levels of inflammatory mediators and cytoprotective proteins in the cells were quantified by enzyme-linked immunosorbent assay and Western blot analysis. RESULTS DHDMP at the concentrations of ≤1 μg/mL did not exhibit a cytotoxic effect for BV2 and HT22 cells. Rather DHDMP effectively restored the growth rate of HT22 cells, which was reduced by co-culture with lipopolysaccharide (LPS)-treated BV2 cells. DHDMP significantly decreased the production of proinflammatory mediators, such as nitric oxide, tumor necrosis factor-α, interleukin-6, inducible nitric oxide synthase, and cyclooxygenase-2 in BV2 cells. Moreover, DHDMP strongly inhibited the nuclear translocation of nuclear factor κB (NF-κB) and phosphorylation of p38 mitogen-activated protein kinase (MAPK) in BV2 cells. The compound did not affect the levels and phosphorylation of ERK and JNK. Concurrently, DHDMP increased the expression of heme oxygenase-1 (HO-1), an inducible cytoprotective enzyme, in HT22 cells. CONCLUSIONS Our findings indicate that DHDMP effectively dampened LPS-mediated inflammatory responses in BV2 microglial cells by suppressing transcriptional activity of NF-κB and its downstream mediators and contributed to HT22 neuronal cell survival. This study provides insight into the therapeutic potential of DHDMP for inflammation-related neurological diseases.
Collapse
Affiliation(s)
- Ji Sun Lim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jisun Oh
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Hyun Seok Yun
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jeong Soon Lee
- Forest Resources Development Institute of Gyeongsangbuk-do, Andong, 36605, South Korea.
| | - Dongyup Hahn
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jong-Sang Kim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea; School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
93
|
do Nascimento RP, dos Santos BL, Amparo JAO, Soares JRP, da Silva KC, Santana MR, Almeida ÁMAN, da Silva VDA, Costa MDFD, Ulrich H, Moura-Neto V, Lopes GPDF, Costa SL. Neuroimmunomodulatory Properties of Flavonoids and Derivates: A Potential Action as Adjuvants for the Treatment of Glioblastoma. Pharmaceutics 2022; 14:pharmaceutics14010116. [PMID: 35057010 PMCID: PMC8778519 DOI: 10.3390/pharmaceutics14010116] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Glioblastomas (GBMs) are tumors that have a high ability to migrate, invade and proliferate in the healthy tissue, what greatly impairs their treatment. These characteristics are associated with the complex microenvironment, formed by the perivascular niche, which is also composed of several stromal cells including astrocytes, microglia, fibroblasts, pericytes and endothelial cells, supporting tumor progression. Further microglia and macrophages associated with GBMs infiltrate the tumor. These innate immune cells are meant to participate in tumor surveillance and eradication, but they become compromised by GBM cells and exploited in the process. In this review we discuss the context of the GBM microenvironment together with the actions of flavonoids, which have attracted scientific attention due to their pharmacological properties as possible anti-tumor agents. Flavonoids act on a variety of signaling pathways, counteracting the invasion process. Luteolin and rutin inhibit NFκB activation, reducing IL-6 production. Fisetin promotes tumor apoptosis, while inhibiting ADAM expression, reducing invasion. Naringenin reduces tumor invasion by down-regulating metalloproteinases expression. Apigenin and rutin induce apoptosis in C6 cells increasing TNFα, while decreasing IL-10 production, denoting a shift from the immunosuppressive Th2 to the Th1 profile. Overall, flavonoids should be further exploited for glioma therapy.
Collapse
Affiliation(s)
- Ravena Pereira do Nascimento
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- Academic College of Nurse, Department of Health, Federal University of Vale do São Francisco, Petrolina 56304-205, Pernambuco, Brazil
| | - Jéssika Alves Oliveira Amparo
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Janaina Ribeiro Pereira Soares
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Karina Costa da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Monique Reis Santana
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Áurea Maria Alves Nunes Almeida
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Victor Diógenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
- Correspondence: (H.U.); (S.L.C.)
| | - Vivaldo Moura-Neto
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Rio de Janeiro, Brazil
- Paulo Niemeyer State Institute of the Brain, Rio de Janeiro 20230-024, Rio de Janeiro, Brazil
| | - Giselle Pinto de Faria Lopes
- Department of Marine Biotechnology, Admiral Paulo Moreira Institute for Sea Studies (IEAPM), Arraial do Cabo 28930-000, Rio de Janeiro, Brazil;
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
- Correspondence: (H.U.); (S.L.C.)
| |
Collapse
|
94
|
Ali EMH, Mersal KI, Ammar UM, Zaraei SO, Abdel-Maksoud MS, El-Gamal MI, Haque MM, Das T, Kim EE, Lee JS, Lee KH, Kim HK, Oh CH. Structural optimization of 4-(imidazol-5-yl)pyridine derivatives affords broad-spectrum anticancer agents with selective B-RAF V600E/p38α kinase inhibitory activity: Synthesis, in vitro assays and in silico study. Eur J Pharm Sci 2022; 171:106115. [PMID: 34995782 DOI: 10.1016/j.ejps.2022.106115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/26/2021] [Accepted: 12/17/2021] [Indexed: 01/10/2023]
Abstract
In the current article, we introduce design of a new series of 4-(imidazol-5-yl)pyridines with improved anticancer activity and selective B-RAFV600E/p38α kinase inhibitory activity. Based on a previous work, a group of structural modifications were applied affording the new potential antiproliferative agents. Towards extensive biological assessment of the target compounds, an in vitro anticancer assay was conducted over NCI 60-cancer cell lines panel representing blood, lung, colon, CNS, skin, ovary, renal, prostate, and breast cancers. Compounds 7c, 7d, 8b, 9b, 9c, 10c, 10d, and 11b exhibited the highest potency among the tested compounds and demonstrated sub-micromolar or one-digit micromolar GI50 values against the majority of the employed cell lines. Compound 10c emerged as the most potent agent with nano-molar activity over most of the cells and incredible activity against melanoma (MDA-MB-435) cell line (GI50 70 nM). It is much more potent than sorafenib, the clinically used anticancer drug, against almost all the NCI-60 cell lines. Further cell-based mechanistic assays showed that compound 10c induced cell cycle arrest and promoted apoptosis in K562, MCF-7 and HT29 cancer cell lines. In addition, compound 10c induced autophagy in the three cancer cell lines. Kinase profiling of 10c showed its inhibitory effects and selectivity towards B-RAFV600E and p38α kinases with IC50 values of 1.84 and 0.726 µM, respectively. Docking of compound 10c disclosed its high affinity in the kinases pockets. Compound 10c represent a promising anticancer agent, that could be optimized in order to improve its kinase activity aiming at developing potential anticancer agents. The conformational stability of compound 10c in the active site of B-RAFV600E and p38α kinases was studied by applying molecular dynamic simulation of the compound in the two kinases for 600 ns in comparison to the native ligands.
Collapse
Affiliation(s)
- Eslam M H Ali
- Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, 12055, Egypt
| | - Karim I Mersal
- Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, 12055, Egypt
| | - Usama M Ammar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0NR, Scotland, United Kingdom
| | - Seyed-Omar Zaraei
- Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea
| | - Mohammed S Abdel-Maksoud
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre NRC (ID: 60014618)), Dokki, Giza, 12622, Egypt
| | - Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Md Mamunul Haque
- Department of Pharmacology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Tanuza Das
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Eunice EunKyeong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jun-Seok Lee
- Department of Pharmacology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Kwan Hyi Lee
- Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hee-Kwon Kim
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Jeonbuk National University Medical School and Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Republic of Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Republic of Korea.
| | - Chang-Hyun Oh
- Center of Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea.
| |
Collapse
|
95
|
Tian Z, Li Z, Guo T, Li H, Mu Y. Atorvastatin suppresses lipopolysaccharide-induced inflammation in human coronary artery endothelial cells. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-979020200001181092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Zhen Tian
- Northeast Agricultural University, China; Harbin Medical University, China
| | | | - Tian Guo
- Harbin Medical University, China
| | - He Li
- Harbin Medical University, China
| | | |
Collapse
|
96
|
de Klerk DJ, de Keijzer MJ, Dias LM, Heemskerk J, de Haan LR, Kleijn TG, Franchi LP, Heger M. Strategies for Improving Photodynamic Therapy Through Pharmacological Modulation of the Immediate Early Stress Response. Methods Mol Biol 2022; 2451:405-480. [PMID: 35505025 DOI: 10.1007/978-1-0716-2099-1_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a minimally to noninvasive treatment modality that has emerged as a promising alternative to conventional cancer treatments. PDT induces hyperoxidative stress and disrupts cellular homeostasis in photosensitized cancer cells, resulting in cell death and ultimately removal of the tumor. However, various survival pathways can be activated in sublethally afflicted cancer cells following PDT. The acute stress response is one of the known survival pathways in PDT, which is activated by reactive oxygen species and signals via ASK-1 (directly) or via TNFR (indirectly). The acute stress response can activate various other survival pathways that may entail antioxidant, pro-inflammatory, angiogenic, and proteotoxic stress responses that culminate in the cancer cell's ability to cope with redox stress and oxidative damage. This review provides an overview of the immediate early stress response in the context of PDT, mechanisms of activation by PDT, and molecular intervention strategies aimed at inhibiting survival signaling and improving PDT outcome.
Collapse
Affiliation(s)
- Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Faculdade de Ciências da Saúde (FCS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Jordi Heemskerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Department of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group, Sciences, and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
97
|
QIAO Y, ZHANG L, HOU C, LI F. Platycodin D protects pancreatic β-cells from STZ-induced oxidative stress and apoptosis. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.63521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yuan QIAO
- Shaanxi Provincial People’s Hospital, China
| | - Lingling ZHANG
- Hospital of Chengdu University of Traditional Chinese Medicine, China
| | | | - Fangzhi LI
- The Fourth Affiliated Hospital of China Medical University, China
| |
Collapse
|
98
|
YIN H, YANG X, LIU S, ZENG J, CHEN S, ZHANG S, LIU Y, ZHAO YT. Total flavonoids from Lagerstroemia speciosa (L.) Pers inhibits TNF-α-induced insulin resistance and inflammatory response in 3T3-L1 adipocytes via MAPK and NF-кB signaling pathways. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.45222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | | | | | | | | | - You LIU
- Ocean University, P. R. China
| | | |
Collapse
|
99
|
Liu Y, Chen L, Liu W, Li D, Zeng J, Tang Q, Zhang Y, Luan F, Zeng N. Cepharanthine Suppresses Herpes Simplex Virus Type 1 Replication Through the Downregulation of the PI3K/Akt and p38 MAPK Signaling Pathways. Front Microbiol 2021; 12:795756. [PMID: 34956164 PMCID: PMC8696181 DOI: 10.3389/fmicb.2021.795756] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cepharanthine (CEP) is a naturally occurring isoquinoline alkaloid extracted from Stephania cepharantha Hayata. Although its underlying molecular mechanism is not fully understood, this compound is reported as a promising antiviral drug. In the present study, we explore the anti-HSV-1 effects and the underlying molecular mechanisms of CEP in vitro. Our results show that CEP could significantly inhibit the formation of plaque and the expression of viral proteins and exhibit a general suppression of replication-associated genes. Whereas HSV-1 infection increases the expressions of phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), and p38 mitogen-activated protein kinase (p38 MAPK) in host cells, CEP was effective indirectly inhibiting phosphorylation levels of the targets in PI3K/Akt and p38 MAPK signaling pathways. Moreover, CEP markedly decreased G0/G1 phase and increased G2/M phase cells and decreased the expression of cyclin-dependent kinase1 (CDK1) and cyclinB1 in a dose-dependent manner. Additionally, CEP increased apoptosis in infected cells, reduced B cell lymphoma-2 (Bcl-2) protein levels, and increased the protein levels of Bcl-associated X protein (Bax), cleaved-caspase3, and nuclear IκB kinaseα (IκBα). Collectively, CEP could arrest the cell cycle in the G2/M phase and induce apoptosis in infected cells by inhibiting the PI3K/Akt and p38 MAPK signaling pathways, hence further reducing HSV-1 infection and subsequent reproduction.
Collapse
Affiliation(s)
- Yao Liu
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Li Chen
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wenjun Liu
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Dan Li
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Jiuseng Zeng
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Tang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuexin Zhang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Fei Luan
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Zeng
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
100
|
Hashimoto T, Kondo N, Hirata M, Temma T. Development of radioiodinated pyrimidinopyridone derivatives as targeted imaging probes of activated p38α for single photon emission computed tomography. Ann Nucl Med 2021; 35:1293-1304. [PMID: 34410619 DOI: 10.1007/s12149-021-01669-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/09/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVE p38α, a member of the mitogen-activated protein kinase superfamily, is ubiquitously expressed in a variety of mammalian cells. Activated p38α induces inflammatory responses to external stimuli, suggesting that non-invasive detection of activated p38α would be valuable for diagnosing inflammatory diseases. For this purpose, we designed radiolabeled compounds [123I]2-IR and [123I]4-IR based on a potent p38α selective inhibitor R1487 for use with single photon emission computed tomography (SPECT). In this study, we used 125I instead of 123I due to its more usable radiochemical properties, synthesized [125I]2-IR and [125I]4-IR, and evaluated their effectiveness as activated p38α imaging probes. METHODS [123I]2-IR and [123I]4-IR were designed by introduction of a 123I atom at the 2- or 4-ositions of the phenoxy ring, preserving the pyrimidinopyridone structure of R1487. We synthesized 2-IR and 4-IR via a 7-step process. The inhibitory potencies of 2-IR, 4-IR, and p38α inhibitors were measured using an ADP-Glo™ kinase assay system. Radioiodination of 2-IR and 4-IR was performed via an organotin-radioiodine exchange reaction using the corresponding tributyltin precursors. Biodistributions were evaluated by determining radioactivity in tissues of interest after intravenous administration of [125I]2-IR and [125I]4-IR in normal ddY mice and turpentine oil-induced inflammation model mice. In vivo inhibition study was also performed in inflammation model mice after intravenous administration of [125I]4-IR with pretreatment of p38α inhibitors. RESULTS We synthesized 2-IR and 4-IR at total yields of 17.5% and 19.2%, respectively. 4-IR had higher p38α inhibitory potency than 2-IR; both compounds were significantly less potent than R1487. [125I]2-IR and [125I]4-IR were successfully obtained from tributyltin precursors with high radiochemical yield (> 65%), purity (> 97%), and molar activity (~ 81 GBq/µmol). [125I]4-IR showed high radioactivity accumulation in the inflamed tissue (7.0 ± 1.2%D/g), rapid delivery throughout the body, and rapid blood clearance, resulting in a high inflammation-to-blood ratio (6.2 ± 0.4) and a high inflammation-to-muscle ratio (5.2 ± 1.3) at 30 min, while [125I]2-IR showed low radioactivity accumulation in inflamed tissue over the experimental period. Further, radioactivity accumulation in inflamed tissue after [125I]4-IR administration was significantly decreased by pretreatment with selective inhibitors. CONCLUSIONS [123I]4-IR would be a promising imaging agent for detection of activated p38α.
Collapse
Affiliation(s)
- Tomoyuki Hashimoto
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Naoya Kondo
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Masahiko Hirata
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Takashi Temma
- Department of Biofunctional Analysis, Graduate School of Pharmaceutical Sciences, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| |
Collapse
|