1
|
Jiang X, Wang W, Kang H. EPHB2 Knockdown Mitigated Myocardial Infarction by Inhibiting MAPK Signaling. Adv Biol (Weinh) 2024; 8:e2300517. [PMID: 38955672 DOI: 10.1002/adbi.202300517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 05/17/2024] [Indexed: 07/04/2024]
Abstract
Myocardial infarction (MI) is a common type of cardiovascular disease. The incidence of ventricular remodeling dysplasia and heart failure increases significantly after MI. The objective of this study is to investigate whether erythropoietin hepatocellular receptor B2 (EPHB2) can regulate myocardial injury after MI and explore its regulatory pathways. EPHB2 is significantly overexpressed in the heart tissues of MI mice. The downregulation of EPHB2 alleviates the cardiac function damage after MI. Knockdown EPHB2 alleviates MI-induced myocardial tissue inflammation and apoptosis, and myocardial fibrosis in mice. EPHB2 knockdown significantly inhibits the activation of mitogen activated kinase-like protein (MAPK) pathway in MI mice. Moreover, EPHB2 overexpression significantly promotes the phosphorylation of MAPK pathway-related protein, which can be reversed by MAPK-IN-1 (an MAPK inhibitor) treatment. In conclusion, silencing EPHB2 can mitigate MI-induced myocardial injury by inhibiting MAPK signaling in mice, suggesting that targeting EPHB2 can be a promising therapeutic target for MI-induced myocardial injury.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Cardiovascular Medicine, Yantai Fushan People's Hospital, Yantai, Shandong, 265500, P. R. China
| | - Wenhua Wang
- Cardiovascular Medicine, Yantai Fushan People's Hospital, Yantai, Shandong, 265500, P. R. China
| | - Haofei Kang
- The First Ward of Cardiovascular Medicine, YanTai YanTaiShan Hospital, Yantai, Shandong, 264000, P. R. China
| |
Collapse
|
2
|
Wang C, Li M, Li S, Wei X, Dong N, Liu S, Yuan Z, Li B, Pierro A, Tang X, Bai Y. Rack1-mediated ferroptosis affects hindgut development in rats with anorectal malformations: Spatial transcriptome insights. Cell Prolif 2024; 57:e13618. [PMID: 38523594 PMCID: PMC11216944 DOI: 10.1111/cpr.13618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 03/26/2024] Open
Abstract
Anorectal malformation (ARM), a common congenital anomaly of the digestive tract, is a result of insufficient elongation of the urorectal septum. The cytoplasmic protein Receptor of Activated C-Kinase 1 (Rack1) is involved in embryonic neural development; however, its role in embryonic digestive tract development and ARM formation is unexplored. Our study explored the hindgut development and cell death mechanisms in ARM-affected rats using spatial transcriptome analysis. We induced ARM in rats by administering ethylenethiourea via gavage on gestational day (GD) 10. On GDs 14-16, embryos from both normal and ARM groups underwent spatial transcriptome sequencing, which identified key genes and signalling pathways. Rack1 exhibited significant interactions among differentially expressed genes on GDs 15 and 16. Reduced Rack1 expression in the ARM-affected hindgut, verified by Rack1 silencing in intestinal epithelial cells, led to increased P38 phosphorylation and activation of the MAPK signalling pathway. The suppression of this pathway downregulated Nqo1 and Gpx4 expression, resulting in elevated intracellular levels of ferrous ions, reactive oxygen species (ROS) and lipid peroxides. Downregulation of Gpx4 expression in the ARM hindgut, coupled with Rack1 co-localisation and consistent mitochondrial morphology, indicated ferroptosis. In summary, Rack1, acting as a hub gene, modulates ferrous ions, lipid peroxides, and ROS via the P38-MAPK/Nqo1/Gpx4 axis. This modulation induces ferroptosis in intestinal epithelial cells, potentially influencing hindgut development during ARM onset.
Collapse
Affiliation(s)
- Chen‐Yi Wang
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Mu‐Yu Li
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Si‐Ying Li
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Xiao‐Gao Wei
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Nai‐Xuan Dong
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Shu‐Ting Liu
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Zheng‐Wei Yuan
- Key Laboratory of Health Ministry for Congenital MalformationShengjing Hospital of China Medical UniversityShenyangChina
| | - Bo Li
- Division of General and Thoracic SurgeryThe Hospital for Sick ChildrenTorontoCanada
| | - Agostino Pierro
- Division of General and Thoracic SurgeryThe Hospital for Sick ChildrenTorontoCanada
| | - Xiao‐Bing Tang
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Yu‐Zuo Bai
- Department of Pediatric SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
3
|
Bris ÁG, MacDowell KS, Ulecia-Morón C, Martín-Hernández D, Moreno B, Madrigal JLM, García-Bueno B, Caso JR, Leza JC. Differential regulation of innate immune system in frontal cortex and hippocampus in a "double-hit" neurodevelopmental model in rats. Neurotherapeutics 2024; 21:e00300. [PMID: 38241165 PMCID: PMC10903097 DOI: 10.1016/j.neurot.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/28/2023] [Indexed: 01/21/2024] Open
Abstract
Neurodevelopmental disorders (NDs) are neuropsychiatric conditions affecting central nervous system development, characterized by cognitive and behavioural alterations. Inflammation has been recently linked to NDs. Animal models are essential for understanding their pathophysiology and identifying therapeutic targets. Double-hit models can reproduce neurodevelopmental and neuroinflammatory impairments. Sixty-seven newborn rats were assigned to four groups: Control, Maternal deprivation (MD, 24-h-deprivation), Isolation (Iso, 5 weeks), and Maternal deprivation + Isolation (MD + Iso, also known as double-hit). Cognitive dysfunction was assessed using behavioural tests. Inflammasome, MAPKs, and TLRs inflammatory elements expression in the frontal cortex (FC) and hippocampus (HP) was analysed through western blot and qRT-PCR. Oxidative/nitrosative (O/N) evaluation and corticosterone levels were measured in plasma samples. Double-hit group was affected in executive and working memory. Most inflammasomes and TLRs inflammatory responses were increased in FC compared to the control group, whilst MAPKs were downregulated. Conversely, hippocampal inflammasome and inflammatory components were reduced after the double-hit exposure, while MAPKs were elevated. Our findings reveal differential regulation of innate immune system components in FC and HP in the double-hit group. Further investigations on MAPKs are necessary to understand their role in regulating HP neuroinflammatory status, potentially linking our MAPKs results to cognitive impairments through their proliferative and anti-inflammatory activity.
Collapse
Affiliation(s)
- Álvaro G Bris
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Karina S MacDowell
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Cristina Ulecia-Morón
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - David Martín-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Beatriz Moreno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - José L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Borja García-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain.
| |
Collapse
|
4
|
Gonzalez L, Sébrié C, Laroche S, Vaillend C, Poirier R. Delayed postnatal brain development and ontogenesis of behavior and cognition in a mouse model of intellectual disability. Neurobiol Dis 2023:106163. [PMID: 37270162 DOI: 10.1016/j.nbd.2023.106163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/05/2023] Open
Abstract
Intellectual disability (ID) is a neurodevelopmental disorder associated with impaired cognitive and adaptive behaviors and represents a major medical issue. Although ID-patients develop behavioral problems and are diagnosed during childhood, most behavioral studies in rodent models have been conducted in adulthood, missing precocious phenotypes expressed during this critical time-window characterized by intense brain plasticity. Here, we selectively assessed postnatal ontogenesis of behavioral and cognitive processes, as well as postnatal brain development in the male Rsk2-knockout mouse model of the Coffin-Lowry syndrome, an X-linked disorder characterized by ID and neurological abnormalities. While Rsk2-knockout mice were born healthy, a longitudinal MRI study revealed a transient secondary microcephaly and a persistent reduction of hippocampal and cerebellar volumes. Specific behavioral parameters from postnatal day 4 (P4) unveiled delayed acquisition of sensory-motor functions and alterations of spontaneous and cognitive behaviors during adolescence, which together, represent hallmarks of neurodevelopmental disorders. Together, our results suggest for the first time that RSK2, an effector of the MAPK signaling pathways, plays a crucial role in brain and cognitive postnatal development. This study also provides new relevant measures to characterize postnatal cognitive development of mouse models of ID and to design early therapeutic approaches.
Collapse
Affiliation(s)
- Laurine Gonzalez
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Catherine Sébrié
- Université Paris-Saclay CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, 91401 Orsay, France
| | - Serge Laroche
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France
| | - Roseline Poirier
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France.
| |
Collapse
|
5
|
Hong E, Min HK, Lim H, Gu SM, Jabborov A, Yayeh T, Kim M, Park WK, Jung JC, Yun J, Oh S. Derivatives of 3, 4, 5-Trimethoxycinnamic Acid Ameliorate Stress-Induced Anxiety in Mice and Rats. Mol Neurobiol 2023; 60:2737-2748. [PMID: 36715919 DOI: 10.1007/s12035-023-03240-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/12/2023] [Indexed: 01/31/2023]
Abstract
Stress is an overwhelming problem associated with neuronal damage leading to anxiety and depression. The compound 3, 4, 5-trimethoxycinnamic acid (TMCA) has shown anti-stress effects; however, its derivatives remained unknown for their anxiolytic properties. Here, therefore, we investigated derivatives of TMCA (dTMCA) for their anxiolytic effects using immobilization and electric shock-induced stress in rats. Derivatives of TMCA ameliorated anxiety in mice and rats revealed by extended period of time spent in the open arms of elevated plus maze. Stress-mediated repression of tyrosine hydroxylase (TH) protein expression in the amygdala regions of rat brain and dopamine levels in the PC12 cells was restored by two selected derivatives (TMCA#5 and TMCA#9). Unlike TH expression, stress-induced protein expression of phospho-extracellular signal-regulated kinase (pERK) was unaffected by both derivatives in rats. Given the preferential inhibitory activity of dTMCA on dopamine and serotonin receptors, serotonergic road map of cellular signaling could be their target for anxiolytic effects. Thus, dTMCA would be promising agents to prevent neuronal damage associated with rampant stressful conditions.
Collapse
Affiliation(s)
- Eunchong Hong
- College of Pharmacy, Chungbuk National University, Cheongju-si, Chungbuk, 28160, Korea
| | - Hyun Kyu Min
- College of Pharmacy, Chungbuk National University, Cheongju-si, Chungbuk, 28160, Korea
| | - Heena Lim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Sun Mi Gu
- College of Pharmacy, Chungbuk National University, Cheongju-si, Chungbuk, 28160, Korea
| | - Abdulaziz Jabborov
- College of Pharmacy, Chungbuk National University, Cheongju-si, Chungbuk, 28160, Korea
| | - Taddesse Yayeh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Mijin Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Woo-Kyu Park
- Medicinal Science Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Korea
| | - Jae-Chul Jung
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, Cheongju-si, Chungbuk, 28160, Korea.
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul, 07804, Korea.
| |
Collapse
|
6
|
Prenatal cyanuric acid exposure disrupts cognitive flexibility and mGluR1-mediated hippocampal long-term depression in male rats. Toxicol Lett 2022; 370:74-84. [PMID: 36152796 DOI: 10.1016/j.toxlet.2022.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/24/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022]
Abstract
Cyanuric acid is one of the most widely used classes of industrial chemicals and is now well known as food adulterant and contaminant in pet food and infant formula. Previously, it was reported that animals prenatally exposed to cyanuric acid showed neurotoxic effects that impaired memory consolidating and suppressed long-term potentiation (LTP) in the hippocampus. However, it is not clear if prenatal exposure to cyanuric acid induces deficits in reversal learning and long-term depression (LTD), which is required for the developmental reorganization of synaptic circuits and updating learned behaviors. Here, pregnant rats were i.p. injected with cyanuric acid (20 mg/kg) during the whole of gestation, and male offspring were selected to examine the levels of hippocampal mGluR1 and mGluR2/3 in young adulthood. The LTD at the Schaffer collateral-CA1 pathway was induced by low-frequency stimulation (LFS) and recorded. Reversal learning and hippocampus-dependent learning strategy were tested in Morris-water maze (MWM) and T-maze tasks, respectively. To further confirm the potential mechanism, selective agonists of mGluR1 and mGluR2/3 and antagonists of mGluR were intra-hippocampal infused before behavioral and neuronal recording. We found the levels of alkaline phosphatase were markedly increased in the maternal placenta and fetal brain following prenatal exposure. The expression of mGluR1 but not mGluR2/3 was significantly decreased and mGluR1-mediated LTD was selectively weakened. Prenatal cyanuric acid impaired reversal learning ability, without changing place learning strategy. The mGluR1 agonist could effectively enhance LFS-induced LTD and mitigate reversal learning deficits. Meanwhile, the reductions in the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPAR)-mediated spontaneous excitatory postsynaptic currents (sEPSCs) amplitude and frequency of cyanuric acid offspring were simultaneously alleviated by mGluR1 agonist infusions. Therefore, the results indicate the cognitive and synaptic impairments induced by prenatal cyanuric acid exposure are attributed to the disruption of the hippocampal mGluR1 signaling. Our findings provided the first evidence for the deteriorated effects of cyanuric acid on synaptic depression and advanced cognitive performance.
Collapse
|
7
|
Li M, Chen L, Zhao Y, Sun H, Zhao L. Research on the Mechanism of HRP Relieving IPEC-J2 Cells Immunological Stress Based on Transcriptome Sequencing Analysis. Front Nutr 2022; 9:944390. [PMID: 35911118 PMCID: PMC9336541 DOI: 10.3389/fnut.2022.944390] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/13/2022] [Indexed: 11/27/2022] Open
Abstract
Early weaning increased the economic benefits of piglets. However, early weaning damages the intestinal barrier of piglets and causes immunological stress. The mechanism by which Hippophae rhamnoides polysaccharide (HRP) alleviates lipopolysaccharide (LPS)-induced intestinal porcine epithelial cells (IPEC-J2) inflammatory damage was investigated using proteomics in our previous studies. In this study we employed RNA-sequencing (RNA-seq) to determine the level and function of differentially expressed genes (DEGs) and further explore the mechanism of the HRP anti-inflammatory and immune process. The differential expression analysis indicated that 3622, 1216, and 2100 DEGs in the IPEC-J2 cells were identified in C vs. L, L vs. H6-L, and C vs. H6-L, respectively. The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis foundsix identified pathways related to the immune system. Additionally, we used the Science, Technology, Engineering, and Math (STEM) program to categorize the 3,134 DEGs that were differentially expressed in H2-L, H4-L and H6-L into eight possible expression profiles, in which 612 were clustered into two profiles. The accuracy and consistency of RNA-seq data were validated by the results of qRT-PCR of the nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (NFKB2), MAP kinase interacting serine/threonine kinase 2 (MKNK2), mitogen-activated protein kinase kinase 1 (MAP2K1), mitogen-activated protein kinase kinase kinase 8 (MAP3K8), Ras-related protein R-Ras (RRAS), TNF receptor-associated factor 1 (TRAF1), NF-kappa-B inhibitor alpha (NFKBIA), interleukin 8 (IL8), tumor necrosis factor, alpha-induced protein 3 (TNFAIP3), and transforming growth factor beta-1 (TGFB1). Transcriptome sequencing also indicated that HRP reduced the expression levels of related DEGs and inhibited the activation of the mitogen-activated protein kinase (MAPK)/nuclear factor kappa-B (NF-κB) signaling pathway. Our findings indicate that the application of HRP in piglet diets during the early weaning period can improve intestinal epithelial function and integrity, and relieve intestinal damage, and improve piglet health.
Collapse
Affiliation(s)
- Muyang Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Lu Chen
- Shanxi Animal Husbandry and Veterinary School, Taiyuan, China
| | - Yiran Zhao
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- *Correspondence: Lei Zhao
| | - Lei Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Hui Sun
| |
Collapse
|
8
|
He D, Hu G, Zhou A, Liu Y, Huang B, Su Y, Wang H, Ye B, He Y, Gao X, Fu S, Liu D. Echinocystic Acid Inhibits Inflammation and Exerts Neuroprotective Effects in MPTP-Induced Parkinson’s Disease Model Mice. Front Pharmacol 2022; 12:787771. [PMID: 35126128 PMCID: PMC8807489 DOI: 10.3389/fphar.2021.787771] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/29/2021] [Indexed: 01/05/2023] Open
Abstract
Parkinson’s disease (PD), the second primary neurodegenerative disease affecting human health, is mainly characterized by dopaminergic neuron damage in the midbrain and the clinical manifestation of movement disorders. Studies have shown that neuroinflammation plays an important role in the progression of PD. Excessively activated microglia produce several pro-inflammatory mediators, leading to damage to the surrounding neurons and finally inducing neurodegeneration. Echinocystic acid (EA) exhibits an anti-inflammatory effect in peripheral tissues. However, whether it inhibited neuroinflammation remains unclear. Therefore, the current study investigates the effect of EA on neuroinflammation and whether it can improve PD symptoms through inhibiting neuroinflammation. In our experiments, we discovered that EA inhibited the production of pro-inflammatory mediators in LPS-exposed BV2 cells. Further mechanism-related studies revealed that EA inhibited inflammation by activating PI3K/Akt and inhibiting NF-κB and MAPK signal pathways in LPS-induced BV2 cells. Research revealed that EA eases microglia-mediated neuron death in SN4741 and SHSY5Y cells. In in vivo studies, the results demonstrated that EA improves weight loss and behavioral impairment in MPTP-induced mice. Further studies have revealed that EA inhibited dopaminergic neuron damage and inflammation in the mice midbrain. In conclusion, our study demonstrated that EA inhibits neuroinflammation and exerts neuroprotective effects by activating PI3K/Akt and inhibiting NF-κB and MAPK signal pathways in vivo and in vitro.
Collapse
Affiliation(s)
- Dewei He
- College of Animal Science, Jilin University, Changchun, China
| | - Guiqiu Hu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ang Zhou
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanting Liu
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Bingxu Huang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yingchun Su
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hefei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Bojian Ye
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuan He
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiyu Gao
- College of Animal Science, Jilin University, Changchun, China
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Dianfeng Liu
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
9
|
Calvo B, Thornton TM, Rincon M, Tranque P, Fernandez M. Regulation of GSK3β by Ser 389 Phosphorylation During Neural Development. Mol Neurobiol 2021; 58:809-820. [PMID: 33029741 DOI: 10.1007/s12035-020-02147-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/22/2020] [Indexed: 10/23/2022]
Abstract
GSK3β is a constitutively active kinase that promotes cell death, which requires strict regulatory mechanisms. Although Akt-mediated phosphorylation at Ser9 is the default mechanism to inactivate GSK3β, phosphorylation of GSK3β at Ser389 by p38 MAPK has emerged as an alternative inhibitory pathway that provides cell protection and repair in response to DNA damage. Phosphorylation of Ser389 GSK3β has been detected in adult brain, where it has been related to neuronal survival and behavior. However, the use of this pathway to regulate GSK3β in the neonatal developing brain is unknown. In this study, we show that phosphorylation of GSK3β at Ser389 in the brain is developmentally regulated, with the highest levels corresponding to the first 2 weeks of age. Moreover, we found that the phosphorylation of GSK3β at Ser389 is the preferential mechanism for inactivating brain GSK3β in 2-week-old mice. Importantly, we show that phospho-Ser389 GSK3β expression is predominant in neuronal cell cultures from neonatal brain relative to other cell populations. However, phospho-Ser389 GSK3β is triggered by DNA double-strand breaks in all developing neural cell types examined. Thus, the phosphorylation of GSK3β on Ser389 could be a central regulatory mechanism to restrain GSK3β during neurogenesis early in life.
Collapse
Affiliation(s)
- Belen Calvo
- Research Institute for Neurological Disabilities (IDINE), Medical School, University of Castilla-La Mancha, 02006, Albacete, Spain
| | - Tina M Thornton
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT, 05405, USA
| | - Mercedes Rincon
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT, 05405, USA
- Department of Immunology and Microbiology, University of Colorado Denver, Aurora, CO, USA
| | - Pedro Tranque
- Research Institute for Neurological Disabilities (IDINE), Medical School, University of Castilla-La Mancha, 02006, Albacete, Spain
| | - Miriam Fernandez
- Research Institute for Neurological Disabilities (IDINE), Medical School, University of Castilla-La Mancha, 02006, Albacete, Spain.
| |
Collapse
|
10
|
Wang X, Li Z, Zhu Y, Yan J, Liu H, Huang G, Li W. Maternal folic acid impacts DNA methylation profile in male rat offspring implicated in neurodevelopment and learning/memory abilities. GENES AND NUTRITION 2021; 16:1. [PMID: 33430764 PMCID: PMC7802276 DOI: 10.1186/s12263-020-00681-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022]
Abstract
Background Periconceptional folic acid (FA) supplementation not only reduces the incidence of neural tube defects, but also improves cognitive performances in offspring. However, the genes or pathways that are epigenetically regulated by FA in neurodevelopment were rarely reported. Methods To elucidate the underlying mechanism, the effect of FA on the methylation profiles in brain tissue of male rat offspring was assessed by methylated DNA immunoprecipitation chip. Differentially methylated genes (DMGs) and gene network analysis were identified using DAVID and KEGG pathway analysis. Results Compared with the folate-normal diet group, 1939 DMGs were identified in the folate-deficient diet group, and 1498 DMGs were identified in the folate-supplemented diet group, among which 298 DMGs were overlapped. The pathways associated with neurodevelopment and learning/memory abilities were differentially methylated in response to maternal FA intake during pregnancy, and there were some identical and distinctive potential mechanisms under FA deficiency or FA-supplemented conditions. Conclusions In conclusion, genes and pathways associated with neurodevelopment and learning/memory abilities were differentially methylated in male rat offspring in response to maternal FA deficiency or supplementation during pregnancy.
Collapse
Affiliation(s)
- Xinyan Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Yun Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China
| | - Jing Yan
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China.,Department of Social Medicine and Health Administration, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | - Huan Liu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, 300070, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, 300070, China. .,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, 300070, China.
| |
Collapse
|
11
|
Luo Z, Yu G, Chen X, Liu Y, Zhou Y, Wang G, Shi Y. Integrated phytochemical analysis based on UHPLC-LTQ-Orbitrap and network pharmacology approaches to explore the potential mechanism of Lycium ruthenicum Murr. for ameliorating Alzheimer's disease. Food Funct 2020; 11:1362-1372. [PMID: 31967149 DOI: 10.1039/c9fo02840d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Based on compelling experimental and clinical evidence, the fruit of Lycium ruthenicum Murr. (LRM), a unique traditional Tibetan medicine, exerts beneficial effects on ameliorating learning and memory deficits of Alzheimer's disease (AD) and other neurodegenerative disorders. However, the potential active constituents and biological mechanism of LRM are still unknown. In this study, the major chemical constituents of LRM were first analyzed by ultra-high-pressure liquid chromatography coupled with linear ion trap-Orbitrap tandem mass spectrometry (UHPLC-LTQ-Orbitrap). A total of 35 constituents were confirmed or tentatively identified. Furthermore, the network-based pharmacological strategy was applied to clarify the molecular mechanism of LRM on AD based on the identified components. Totally, 143 major targets were screened and supposed to be effective players in alleviating AD. Then, the LRM chemicals-major LRM putative targets-major pathways network was constructed, implying potential biological function of LRM on AD. More importantly, 12 core genes which can be modulated by LRM were identified, and they may play a pivotal role in alleviating some major symptoms of AD. This study provided a scientific basis for further investigation and application of LRM, which demonstrated that the network pharmacology approach could be a powerful way for the mechanistic studies of folk medicines.
Collapse
Affiliation(s)
- Zhiqiang Luo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China. and School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Guohua Yu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xinjing Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yating Zhou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Guopeng Wang
- Zhongcai Health (Beijing) Biological Technology Development Co., Ltd., Beijing 101500, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
12
|
Sajanti A, Lyne SB, Girard R, Frantzén J, Rantamäki T, Heino I, Cao Y, Diniz C, Umemori J, Li Y, Takala R, Posti JP, Roine S, Koskimäki F, Rahi M, Rinne J, Castrén E, Koskimäki J. A comprehensive p75 neurotrophin receptor gene network and pathway analyses identifying new target genes. Sci Rep 2020; 10:14984. [PMID: 32917932 PMCID: PMC7486379 DOI: 10.1038/s41598-020-72061-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022] Open
Abstract
P75 neurotrophic receptor (p75NTR) is an important receptor for the role of neurotrophins in modulating brain plasticity and apoptosis. The current understanding of the role of p75NTR in cellular adaptation following pathological insults remains blurred, which makes p75NTR’s related signaling networks an interesting and challenging initial point of investigation. We identified p75NTR and related genes through extensive data mining of a PubMed literature search including published works related to p75NTR from the past 20 years. Bioinformatic network and pathway analyses of identified genes (n = 235) were performed using ReactomeFIViz in Cytoscape based on the highly reliable Reactome functional interaction network algorithm. This approach merges interactions extracted from human curated pathways with predicted interactions from machine learning. Genome-wide pathway analysis showed total of 16 enriched hierarchical clusters. A total of 278 enriched single pathways were also identified (p < 0.05, false discovery rate corrected). Gene network analyses showed multiple known and new targets in the p75NTR gene network. This study provides a comprehensive analysis and investigation into the current knowledge of p75NTR signaling networks and pathways. These results also identify several genes and their respective protein products as involved in the p75NTR network, which have not previously been clearly studied in this pathway. These results can be used to generate novel hypotheses to gain a greater understanding of p75NTR in acute brain injuries, neurodegenerative diseases and general response to cellular damage.
Collapse
Affiliation(s)
- Antti Sajanti
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Seán B Lyne
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Janek Frantzén
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences and Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Iiro Heino
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Cassiano Diniz
- Neuroscience Center, HiLIFE, University of Helsinki, Box 63, 00014, Helsinki, Finland
| | - Juzoh Umemori
- Neuroscience Center, HiLIFE, University of Helsinki, Box 63, 00014, Helsinki, Finland
| | - Yan Li
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA.,Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Riikka Takala
- Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, POB 52, 20521, Turku, Finland.,Department of Anaesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Jussi P Posti
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Susanna Roine
- Division of Clinical Neurosciences, Department of Cerebrovascular Diseases, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Fredrika Koskimäki
- Division of Clinical Neurosciences, Department of Cerebrovascular Diseases, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Melissa Rahi
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Jaakko Rinne
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland
| | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, Box 63, 00014, Helsinki, Finland
| | - Janne Koskimäki
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital and University of Turku, Hämeentie 11, P.O. Box 52, 20521, Turku, Finland. .,Department of Psychiatry, Central Hospital of Southern Ostrobothnia, Hanneksenrinne 7, 60220, Seinäjoki, Finland.
| |
Collapse
|
13
|
Lai J, Peng L, Chen H, Li R, Liu L, Liu J, Shi X. Amino acid sequence identification and anti-inflammatory potency evaluation of dandelion oligopeptides in LPS-induced RAW264.7 macrophage cells. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
14
|
Albert-Gascó H, Ros-Bernal F, Castillo-Gómez E, Olucha-Bordonau FE. MAP/ERK Signaling in Developing Cognitive and Emotional Function and Its Effect on Pathological and Neurodegenerative Processes. Int J Mol Sci 2020; 21:E4471. [PMID: 32586047 PMCID: PMC7352860 DOI: 10.3390/ijms21124471] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The signaling pathway of the microtubule-associated protein kinase or extracellular regulated kinase (MAPK/ERK) is a common mechanism of extracellular information transduction from extracellular stimuli to the intracellular space. The transduction of information leads to changes in the ongoing metabolic pathways and the modification of gene expression patterns. In the central nervous system, ERK is expressed ubiquitously, both temporally and spatially. As for the temporal ubiquity, this signaling system participates in three key moments: (i) Embryonic development; (ii) the early postnatal period; and iii) adulthood. During embryonic development, the system is partly responsible for the patterning of segmentation in the encephalic vesicle through the FGF8-ERK pathway. In addition, during this period, ERK directs neurogenesis migration and the final fate of neural progenitors. During the early postnatal period, ERK participates in the maturation process of dendritic trees and synaptogenesis. During adulthood, ERK participates in social and emotional behavior and memory processes, including long-term potentiation. Alterations in mechanisms related to ERK are associated with different pathological outcomes. Genetic alterations in any component of the ERK pathway result in pathologies associated with neural crest derivatives and mental dysfunctions associated with autism spectrum disorders. The MAP-ERK pathway is a key element of the neuroinflammatory pathway triggered by glial cells during the development of neurodegenerative diseases, such as Parkinson's and Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, as well as prionic diseases. The process triggered by MAPK/ERK activation depends on the stage of development (mature or senescence), the type of cellular element in which the pathway is activated, and the anatomic neural structure. However, extensive gaps exist with regards to the targets of the phosphorylated ERK in many of these processes.
Collapse
Affiliation(s)
- Héctor Albert-Gascó
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Hills Road, Cambridge CB2 0AH, UK;
| | - Francisco Ros-Bernal
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
| | - Esther Castillo-Gómez
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| | - Francisco E. Olucha-Bordonau
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| |
Collapse
|
15
|
Shcherbitskaia AD, Vasilev DS, Milyutina YP, Tumanova NL, Zalozniaia IV, Kerkeshko GO, Arutjunyan AV. Maternal Hyperhomocysteinemia Induces Neuroinflammation and Neuronal Death in the Rat Offspring Cortex. Neurotox Res 2020; 38:408-420. [PMID: 32504390 DOI: 10.1007/s12640-020-00233-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/19/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Maternal hyperhomocysteinemia is one of the common complications of pregnancy that causes offspring cognitive deficits during postnatal development. In the present work, we evaluated the effect of prenatal hyperhomocysteinemia on structural and ultrastructural organization, neuronal and glial cell number, apoptosis (caspase-3 content and activity), inflammatory markers (tumor necrosis factor-α, interleukin-6, and interleukin-1β), and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation in the offspring brain cortex in early ontogenesis. Wistar female rats received methionine (0.6 g/kg body weight) by oral administration during pregnancy. Histological and biochemical analyses of 5- and 20-day-old pups' cortical tissue were performed. Lysosome accumulation and other neurodegenerative changes in neurons of animals with impaired embryonic development were investigated by electron microscopy. Neuronal staining (anti-NeuN) revealed a reduction in neuronal number, accompanied by increasing of caspase-3 active form protein level and activity. Maternal hyperhomocysteinemia also elevated the number of astroglial and microglial cells and increased expression of interleukin-1β and p38 MAPK phosphorylation, which indicates the development of neuroinflammatory processes.
Collapse
Affiliation(s)
- A D Shcherbitskaia
- D.O. Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, Russia. .,I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia.
| | - D S Vasilev
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Yu P Milyutina
- D.O. Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, Russia
| | - N L Tumanova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - I V Zalozniaia
- D.O. Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, Russia
| | - G O Kerkeshko
- Saint Petersburg Institute of Bioregulation and Gerontology, St. Petersburg, Russia
| | - A V Arutjunyan
- D.O. Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, Russia
| |
Collapse
|
16
|
Li Y, Liu L, Sun P, Zhang Y, Wu T, Sun H, Cheng KW, Chen F. Fucoxanthinol from the Diatom Nitzschia Laevis Ameliorates Neuroinflammatory Responses in Lipopolysaccharide-Stimulated BV-2 Microglia. Mar Drugs 2020; 18:E116. [PMID: 32079242 PMCID: PMC7074591 DOI: 10.3390/md18020116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/09/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, microalgae have drawn increasing attention as a valuable source of functional food ingredients. Intriguingly, Nitzschia laevis is rich in fucoxanthinol that is seldom found in natural sources. Fucoxanthinol, a marine xanthophyll carotenoid, possesses various beneficial bioactivities. Nevertheless, it's not clear whether fucoxanthinol could exert anti-neuroinflammatory function. In light of these premises, the aim of the present study was to investigate the anti-inflammatory role of fucoxanthinol purified from Nitzschia laevis in Lipopolysaccharide (LPS)-stimulated microglia. The results showed that pre-treatment of fucoxanthinol remarkably attenuated the expression of LPS-induced nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), and the production of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), prostaglandin E2 (PGE-2), nitric oxide (NO) and reactive oxygen species (ROS) induction. Modulation mechanism studies revealed that fucoxanthinol hampered nuclear factor-kappa B (NF-κB), Akt, and mitogen-activated protein kinase (MAPK) pathways. Meanwhile, fucoxanthinol led to the enhancement of nuclear translocation of NF-E2-related factor 2 (Nrf2), and the upregulation of heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO-1). Taken together, the results indicated that fucoxanthinol obtained from Nitzschia laevis had great potential as a neuroprotective agent in neuroinflammation and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuelian Li
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing 100871, China; (Y.L.)
| | - Lu Liu
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing 100871, China; (Y.L.)
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Peipei Sun
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing 100871, China; (Y.L.)
| | - Yifeng Zhang
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing 100871, China; (Y.L.)
| | - Tao Wu
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing 100871, China; (Y.L.)
| | - Han Sun
- Institute for Food and Bioresource Engineering, College of Engineering, Peking University, Beijing 100871, China; (Y.L.)
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Ka-Wing Cheng
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
17
|
Yang C, Yan Z, Hu F, Wei W, Sun Z, Xu W. Silencing of microRNA-517a induces oxidative stress injury in melanoma cells via inactivation of the JNK signaling pathway by upregulating CDKN1C. Cancer Cell Int 2020; 20:32. [PMID: 32015692 PMCID: PMC6990552 DOI: 10.1186/s12935-019-1064-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 12/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background Melanoma is notoriously resistant to current treatments, and less than 25% of metastatic melanoma cases respond to existing therapies. Growing evidence has shown that microRNAs (miRNAs) play a vital role in the prognosis of melanoma. MiR-517a has been implicated in many types of cancer; however, its expressional features and potential biological functions in melanoma remain unclear. The present study aimed to investigate the possible effects of miR-517a on oxidative stress (OS) in melanoma cells. Methods miR-517a expression in melanoma was determined using RT-qPCR. After treatment with different concentrations of H2O2, cell viability was determined in order to identify the most appropriate H2O2 concentration. Through loss and gain of function experiments, the interactions between miR-517a, the cyclin dependent kinase inhibitor 1C (CDKN1C) and the c-Jun NH2-terminal kinase (JNK) signaling pathway, as well as their roles in OS of melanoma cells were identified. Moreover, the expression of Cleaved Caspase-3, extent of ERK1/2 phosphorylation, Bax/Bcl-2 ratio, levels of T-AOC, ROS and MDA, and SOD activity were also tested. Finally, melanoma cell viability and apoptosis were detected. Results MiR-517a was upregulated, while CDKN1C was downregulated in melanoma tissues and cells. MiR-517a targets CDKN1C and consequently reduced its expression. Inhibition of miR-517a was shown to increase Cleaved Caspase-3 expression, Bax/Bcl-2 ratio, levels of ROS and MDA, as well as cell apoptosis but decrease extent of ERK1/2 phosphorylation, T-AOC levels, SOD activity, along with cell proliferation and mitochondrial membrane potential. Conclusions Overall, silencing miR-517a results in upregulated CDKN1C expression, and inhibited JNK signaling pathway activation, consequently promoting OS in melanoma cells.
Collapse
Affiliation(s)
- Chao Yang
- 1Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021 Hubei People's Republic of China
| | - Zeqiang Yan
- 2Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021 People's Republic of China
| | - Fen Hu
- 1Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021 Hubei People's Republic of China
| | - Wei Wei
- 1Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021 Hubei People's Republic of China
| | - Zhihua Sun
- 1Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021 Hubei People's Republic of China
| | - Wei Xu
- 3Department of Dermatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021 Hubei People's Republic of China
| |
Collapse
|
18
|
Yang H, Wang Q, Han L, Yang X, Zhao W, Lyu L, Wang L, Yan H, Che C. Nerolidol inhibits the LOX-1 / IL-1β signaling to protect against the Aspergillus fumigatus keratitis inflammation damage to the cornea. Int Immunopharmacol 2020; 80:106118. [PMID: 31926445 DOI: 10.1016/j.intimp.2019.106118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/01/2019] [Accepted: 12/07/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Nerolidol, a naturally occurring sesquiterpene has both anti-microbial and anti-inflammatory properties. The current study aims to investigate the antifungal and the anti-inflammatory effects of nerolidol against mouse Aspergillus fumigatus (A. fumigatus) keratitis. METHODS The minimum inhibitory concentration (MIC) and cytotoxicity tests were used to study the antifungal ability. For in vivo and in vitro studies, the mouse corneas and the human corneal epithelial cells (HCECs) infected with A. fumigatus spores were intervented with nerolidol or phosphate buffer saline (PBS). Thereafter, the effect of the nerolidol on the response against inflammation was analyzed using the following parameters: recruitment of the neutrophils or macrophages and the expression of the lectin-type oxidized low density lipoprotein receptor-1 (LOX-1) and interleukin 1β (IL-1β). Techniques used were the slit lamp, immunofluorescence, myeloperoxidase (MPO) detection, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. RESULTS Nerolidol directly inhibits the growth of A. fumigatus. The administration of nerolidol reduced the severity of fungal keratitis with infiltration of fewer inflammatory cells and reduced levels of the LOX-1, as well the anti-inflammatory cytokines such as IL-1β were reduced compared with the PBS group. Additionally, in vitro studies showed that treatment with nerolidol inhibited the production of the LOX-1 / IL-1β levels in A. fumigatus stimulated HCECs. CONCLUSION Nerolidol attenuated the A. fumigatus keratitis inflammatory response by inhibiting the growth of A. fumigatus, reducing the recruitment of the neutrophils and the macrophages, and inhibiting the LOX-1/ IL-1β signaling.
Collapse
Affiliation(s)
- Hua Yang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qian Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Lin Han
- Gout Laboratory, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xuejiao Yang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Wenyi Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Leyu Lyu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Limei Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Haijing Yan
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chengye Che
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
19
|
Gu F, Wu Y, Liu Y, Dou M, Jiang Y, Liang H. Lactobacillus casei improves depression-like behavior in chronic unpredictable mild stress-induced rats by the BDNF-TrkB signal pathway and the intestinal microbiota. Food Funct 2020; 11:6148-6157. [DOI: 10.1039/d0fo00373e] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
L. casei improves depression-like behavior in stress-induced rats by the BDNF-TrkB signal pathway and the intestinal microbiota.
Collapse
Affiliation(s)
- Fang Gu
- College of Mechanical and Electronic Engineering
- Northwest A&F University
- Yangling 712100
- China
| | - Yanyan Wu
- Department of Human Nutrition
- College of Public Health
- Qingdao University
- Qingdao 266071
- China
| | - Ying Liu
- College of Basic Medicine
- Qingdao University
- Qingdao 266071
- China
| | - Mei Dou
- Department of Human Nutrition
- College of Public Health
- Qingdao University
- Qingdao 266071
- China
| | - Yushan Jiang
- Department of Human Nutrition
- College of Public Health
- Qingdao University
- Qingdao 266071
- China
| | - Hui Liang
- Department of Human Nutrition
- College of Public Health
- Qingdao University
- Qingdao 266071
- China
| |
Collapse
|
20
|
Chen MQ, Luan JJ. HMGB1 promotes bone fracture healing through activation of ERK signaling pathway in a rat tibial fracture model. Kaohsiung J Med Sci 2019; 35:550-558. [PMID: 31162822 DOI: 10.1002/kjm2.12095] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 05/12/2019] [Indexed: 01/04/2023] Open
Abstract
This work was to investigate potential roles of HMGB1-mediated ERK pathway in the healing process of bone fracture. Rat tibial fracture models were established and divided into control (rats with normal saline), HMGB1 (rats with HMGB1), and HMGB1+ PD98059 groups (rats with HMGB1 and 1 mg/kg of ERK1/2 inhibitor PD98059) with 30 rats per each. The healing of rats' fracture was observed by X-ray films, the morphological changes of bone fractures by HE staining, the callus formation by micro-CT and biomechanical test, and the expression of osteogenesis-related genes, HMGB1 and ERK-related proteins by qRT-PCR and Western blot. Rats in the HMGB1 group was increased in X-ray scores, peak torque, torsional stiffness, and the bone volume fraction (bone volume/total volume, BV/TV); meanwhile, those rats presented elevations in osteogenesis-related genes and HMGB1 expressions, as well as p-ERK/ERK ratio. However, rats in the HMGB1+ PD98059 group was significantly reduced in X-ray score, peak torque, torsional stiffness, and BV/TV, as well as the expression of osteogenesis-related genes and the ratio of p-ERK/ERK, as compared to those from HMGB1 group. HMGB1 could promote the expressions of osteogenesis-related genes and accelerate the healing process of fracture via activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Ming-Qi Chen
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| | - Jing-Jie Luan
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| |
Collapse
|
21
|
Upregulation of long noncoding RNA Gadd45a is associated with sevoflurane-induced neurotoxicity in rat neural stem cells. Neuroreport 2019. [PMID: 29521679 DOI: 10.1097/wnr.0000000000000980] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Emerging evidence has shown that long noncoding RNA (lncRNA) plays a crucial role in controlling neural stem cells' (NSCs) survival. However, the fundamental role of lncRNA underlying sevoflurane-induced neurotoxicity remains poorly elucidated. In the present study, we investigate the effect of sevoflurane-induced neurotoxicity in a concentration-dependent and duration-dependent manner. Furthermore, we assayed the differential profile of lncRNA in rat hippocampal NSCs following sevoflurane exposure, and identified lncRNA Gadd45a and the correlation between lncRNA Gadd45a and Gadd45a. We found that lncRNA Gadd45a and its nearby gene, Gadd45a, were significantly upregulated in NSCs exposed to sevoflurane. Notably, Gadd45a was enriched in the cell cycle-relative pathway including mitogen-activated protein kinases and P53 signaling, whereas lncRNA Gadd45a was positively correlated with Gadd45a. These results suggest lncRNA Gadd45a is associated with sevoflurane-induced toxicity, and thus shed light on a new key target for revealing the molecular mechanism of sevoflurane-induced toxicity.
Collapse
|
22
|
Ke T, Gonçalves FM, Gonçalves CL, Dos Santos AA, Rocha JBT, Farina M, Skalny A, Tsatsakis A, Bowman AB, Aschner M. Post-translational modifications in MeHg-induced neurotoxicity. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2068-2081. [PMID: 30385410 DOI: 10.1016/j.bbadis.2018.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Mercury (Hg) exposure remains a major public health concern due to its widespread distribution in the environment. Organic mercurials, such as MeHg, have been extensively investigated especially because of their congenital effects. In this context, studies on the molecular mechanism of MeHg-induced neurotoxicity are pivotal to the understanding of its toxic effects and the development of preventive measures. Post-translational modifications (PTMs) of proteins, such as phosphorylation, ubiquitination, and acetylation are essential for the proper function of proteins and play important roles in the regulation of cellular homeostasis. The rapid and transient nature of many PTMs allows efficient signal transduction in response to stress. This review summarizes the current knowledge of PTMs in MeHg-induced neurotoxicity, including the most commonly PTMs, as well as PTMs induced by oxidative stress and PTMs of antioxidant proteins. Though PTMs represent an important molecular mechanism for maintaining cellular homeostasis and are involved in the neurotoxic effects of MeHg, we are far from understanding the complete picture on their role, and further research is warranted to increase our knowledge of PTMs in MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Cinara Ludvig Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | | | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, 97105900 Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040900 Florianópolis, SC, Brazil
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
23
|
Cao Y, Pan T, Chen X, Wu J, Guo N, Wang B. EP4 knockdown alleviates glomerulosclerosis through Smad and MAPK pathways in mesangial cells. Mol Med Rep 2018; 18:5141-5150. [PMID: 30320390 DOI: 10.3892/mmr.2018.9553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 09/21/2018] [Indexed: 11/06/2022] Open
Abstract
Prostaglandin E2 has exhibited pleiotropic effects in the regulation of glomerulosclerosis progression through its four receptors. The current study aimed to evaluate the effect of prostaglandin receptor EP4 on mesangial cell proliferation. In vivo, 5/6 nephrectomy was introduced into EP4+/‑ and wild‑type (WT) mice. Clinical parameters were monitored post‑surgery. At 8 weeks post‑surgery, glomerular fibrosis‑associated indicators were measured by immunohistochemical staining and trichrome staining. In vitro, mesangial cells in different groups (transfected with green fluorescent protein, AD‑EF4 or AD‑CRE) were exposed to transforming growth factor (TGF)‑β1 for 24 h to detect the level of downstream signaling. Corresponding signaling inhibitors were also used to validate the signaling effects. Following surgery, EP4+/‑ mice presented a higher survival rate and normal urine volume compared with the WT group, and serum creatinine level and 24 h urine protein were lower in the EP4+/‑ mice. Furthermore, associated profibrotic indicators were identified to have decreased at 8 weeks post‑surgery along with less tubule‑interstitial fibrosis. In vivo, the inhibition of extracellular signal‑regulated kinase and P38 phosphorylation alleviated the accumulation of mesangial matrix, and these signals were enhanced when EP4 was overexpressed. EP4 enhancement aggravated imbalanced mesangial cell proliferation stimulated by TGF‑β1 and GS of mice treated with 5/6 nephrectomy through the Smad and mitogen‑activated protein kinase pathways.
Collapse
Affiliation(s)
- Yingjie Cao
- Department of Nephrology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Tianyi Pan
- Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200000, P.R. China
| | - Xiaolan Chen
- Department of Nephrology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Jianhua Wu
- Department of Nephrology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Naifeng Guo
- Department of Nephrology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Bicheng Wang
- Basic Medical College, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
24
|
Zhao C, Zhao J, Yang Q, Ye Y. Cobra neurotoxin produces central analgesic and hyperalgesic actions via adenosine A 1 and A 2A receptors. Mol Pain 2018; 13:1744806917720336. [PMID: 28758541 PMCID: PMC5542074 DOI: 10.1177/1744806917720336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cobra neurotoxin, a short-chain peptide isolated from snake venom of Naja naja atra, showed both a central analgesic effect and a hyperalgesic effect in mice tests. In order to explore mechanisms, a hypothesis is put forward that cobra neurotoxin takes effect through adenosine receptor pathway. The central effects of cobra neurotoxin were evaluated using the hot plate test (a model of acute pain) and the spinal cord injury (a model of central pain) in mice and using A1 receptor antagonist (DPCPX) and A2A receptor antagonist (ZM241385); behaviors were scored and signal molecules such as reactive oxygen species and adenosine triphosphate levels and mitogen-activated protein kinases/extracellular signal-regulated protein kinase expression were measured. Low dose of cobra neurotoxin (25 µg/kg) had analgesic effects which were inhibited by DPCPX, while high dose of cobra neurotoxin (100 µg/kg) had hyperalgesic effects which were blocked by ZM241385. Cobra neurotoxin reduced reactive oxygen species and increased adenosine triphosphate in brain tissues, and extracellular signal-regulated protein kinase expression was markedly inhibited by cobra neurotoxin. Cobra neurotoxin may take effect through mitogen-activated protein kinases/extracellular signal-regulated protein kinase pathway inhibition by activating adenosine A1Rs and cause changes of reactive oxygen species and adenosine triphosphate through feedback mechanisms. Overdose cobra neurotoxin further activates the adenosine A2ARs to generate pain sensitization. This research proposes a new central analgesic mechanism of cobra neurotoxin and discloses dual regulation of pain.
Collapse
Affiliation(s)
- Chuang Zhao
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, China
| | - Jun Zhao
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, China
| | - Qian Yang
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, China
| | - Yong Ye
- Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, China
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, Guangzhou, China
- Yong Ye, Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
25
|
Zhang B, Li SL, Xie HL, Fan JW, Gu CW, Kang C, Teng MJ. Effects of silencing the DUSP1 gene using lentiviral vector-mediated siRNA on the release of proinflammatory cytokines through regulation of the MAPK signaling pathway in mice with acute pancreatitis. Int J Mol Med 2018; 41:2213-2224. [PMID: 29393354 DOI: 10.3892/ijmm.2018.3429] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/10/2018] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the effects of dual specificity phosphatase 1 (DUSP1) gene silencing using lentiviral vector-mediated small interfering (si)RNA on the release of proinflammatory cytokines through the regulation of the mitogen‑activated protein kinase (MAPK) signaling pathway in mice with acute pancreatitis (AP). Two siRNA‑DUSP1 sequences and one scramble siRNA sequence were designed, and the expression of DUSP1 was detected using western blot analysis to screen for the one with a higher interference rate. An AP mouse model was established, and KM mice were assigned to either a control, siRNA, AP, AP+PD98059, AP+scramble, AP+siRNA or AP+PD98059+siRNA group. The expression of proinflammatory cytokines, including tumor necrosis factor (TNF)‑α, interleukin (IL)‑1β and IL‑6, high mobility group box 1 (HMGB1), and S100A12 in serum samples were detected using an enzyme‑linked immunosorbent assay at 12, 24 and 48 h post‑modeling. The serum amylase levels were also detected. The expression levels of DUSP1, TNF‑α, IL‑1β, IL‑6, HMGB1, S100A12, phosphorylated (p‑) extracellular signal‑regulated kinase (ERK), p‑c‑Jun N‑terminal kinase (JNK), p‑p38, ERK, JNK and p38 in pancreatic, liver, kidney and lung tissues were detected using reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Compared with the control group, the siRNA group demonstrated marginally upregulated serum amylase, lipase, urinary trypsinogen‑2, and proinflammatory cytokines, HMGB1 and S100A12 in serum and tissues, with no statistically significant difference, elevated expression levels of p‑ERK, p‑JNK and p‑p38, and decreased expression of DUSP1. The other five groups demonstrated increased expression levels of TNF‑α, IL‑1β, IL‑6, HMGB1, S100A12, amylase, lipase and urinary trypsinogen‑2 in serum, and increased expression levels of DUSP1, TNF‑α, IL‑1β, IL‑6, HMGB1, S100A12, p‑ERK, p‑JNK and p‑p38 in tissues. Compared with the AP group, the AP+PD98059+siRNA group had decreased expression of DUSP1 in tissues, whereas the AP+PD98059 group had decreased serum expression levels of TNF‑α, IL‑1β, IL‑6, HMGB1, S100A12 and amylase, lipase and urinary trypsinogen‑2. The expression levels of TNF‑α, IL‑1β, IL‑6, HMGB1, S100A12, p‑ERK, p‑JNK, p‑p38 in tissues, and edema of pancreatic tissue were alleviated, whereas the opposite results were observed in the AP+siRNA group with the decreased expression of DUSP1. The results suggested that DUSP1 gene silencing promoted the release of proinflammatory cytokines through activation of the MAPK signaling pathway in mice with AP.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Hepatobiliary Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Shu-Liang Li
- Department of General Surgery, The Second People's Hospital of Liaocheng, Liaocheng, Shandong 252600, P.R. China
| | - Hua-Lei Xie
- Department of Emergency, The Second People's Hospital of Liaocheng, Liaocheng, Shandong 252600, P.R. China
| | - Jia-Wei Fan
- Department of Emergency, The Second People's Hospital of Liaocheng, Liaocheng, Shandong 252600, P.R. China
| | - Chuan-Wei Gu
- Department of Emergency, The Second People's Hospital of Liaocheng, Liaocheng, Shandong 252600, P.R. China
| | - Chao Kang
- Department of Emergency, The Second People's Hospital of Liaocheng, Liaocheng, Shandong 252600, P.R. China
| | - Mu-Jian Teng
- Department of Hepatobiliary Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
26
|
Yuan K, Zhao G, Che C, Li C, Lin J, Zhu G, He K. Dectin-1 is essential for IL-1β production through JNK activation and apoptosis in Aspergillus fumigatus keratitis. Int Immunopharmacol 2017; 52:168-175. [PMID: 28926773 DOI: 10.1016/j.intimp.2017.09.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE To investigate the role of phosphorylated JNK in Dectin-1-induced IL-1β production and the role of Dectin-1 in apoptosis in mouse Aspergillus fumigatus (A. fumigatus) keratitis. METHODS Mice corneas were pretreated with Dectin-1 siRNA or SP600125 (the inhibitor of JNK) before A. fumigatus infection. THP-1 macrophages were preincubated with SP600125 before the stimulation of A. fumigatus conidia. Dectin-1, IL-1β, JNK, Bax, Bcl-2, cytochrome-c (cyt-c), caspase-9, caspase-8 and caspase-3 expressions were tested by PCR, Western blot, or Immunofluorescence staining. RESULTS Pretreatment with Dectin-1 siRNA significantly decreased A. fumigatus-induced IL-1β production and JNK phosphorylation compared with scrambled control in C57BL/6 mice corneas. SP600125 treatment before infection significantly inhibited IL-1β production compared with DMSO control both in mice corneas and THP-1 macrophages. Furthermore, Dectin-1 deficiency resulted in increased ratio of Bax/Bcl-2, release of cyt-c, activation of caspase-9 and caspase-3 in mouse A. fumigatus keratitis. However, Dectin-1 deficiency didn't affect the activation of caspase-8. CONCLUSIONS Being an important inflammatory PRR to mediate host inflammatory response, Dectin-1 induced IL-1β production is JNK dependent in mouse A. fumigatus keratitis, and suppressed apoptosis mediated anti-inflammatory response.
Collapse
Affiliation(s)
- Kelan Yuan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China.
| | - Chengye Che
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guoqiang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Kun He
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
27
|
Lopes MW, Leal RB, Guarnieri R, Schwarzbold ML, Hoeller A, Diaz AP, Boos GL, Lin K, Linhares MN, Nunes JC, Quevedo J, Bortolotto ZA, Markowitsch HJ, Lightman SL, Walz R. A single high dose of dexamethasone affects the phosphorylation state of glutamate AMPA receptors in the human limbic system. Transl Psychiatry 2016; 6:e986. [PMID: 27959333 PMCID: PMC5290343 DOI: 10.1038/tp.2016.251] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/15/2016] [Indexed: 12/28/2022] Open
Abstract
Glucocorticoids (GC) released during stress response exert feedforward effects in the whole brain, but particularly in the limbic circuits that modulates cognition, emotion and behavior. GC are the most commonly prescribed anti-inflammatory and immunosuppressant medication worldwide and pharmacological GC treatment has been paralleled by the high incidence of acute and chronic neuropsychiatric side effects, which reinforces the brain sensitivity for GC. Synapses can be bi-directionally modifiable via potentiation (long-term potentiation, LTP) or depotentiation (long-term depression, LTD) of synaptic transmission efficacy, and the phosphorylation state of Ser831 and Ser845 sites, in the GluA1 subunit of the glutamate AMPA receptors, are a critical event for these synaptic neuroplasticity events. Through a quasi-randomized controlled study, we show that a single high dexamethasone dose significantly reduces in a dose-dependent manner the levels of GluA1-Ser831 phosphorylation in the amygdala resected during surgery for temporal lobe epilepsy. This is the first report demonstrating GC effects on key markers of synaptic neuroplasticity in the human limbic system. The results contribute to understanding how GC affects the human brain under physiologic and pharmacologic conditions.
Collapse
Affiliation(s)
- M W Lopes
- Department of Biochemistry, Federal University of Santa Catarina, Floranópolis, Brazil
| | - R B Leal
- Department of Biochemistry, Federal University of Santa Catarina, Floranópolis, Brazil,Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil
| | - R Guarnieri
- Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil,Epilepsy Center of Santa Catarina, Federal University of Santa Catarina, Florianópolis, Brazil,Neurosurgery Unit, Governador Celso Ramos Hospital, Florianópolis, Brazil
| | - M L Schwarzbold
- Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil,Department of Internal Medicine, Federal University of Santa Catarina, Florianópolis, Brazil
| | - A Hoeller
- Department of Biochemistry, Federal University of Santa Catarina, Floranópolis, Brazil
| | - A P Diaz
- Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil,Department of Internal Medicine, Federal University of Santa Catarina, Florianópolis, Brazil
| | - G L Boos
- Anesthesiology Division, Hospital Governador Celso Ramos, Florianópolis, Brazil
| | - K Lin
- Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil,Epilepsy Center of Santa Catarina, Federal University of Santa Catarina, Florianópolis, Brazil,Department of Internal Medicine, Federal University of Santa Catarina, Florianópolis, Brazil
| | - M N Linhares
- Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil,Neurosurgery Unit, Governador Celso Ramos Hospital, Florianópolis, Brazil,Department of Surgery, HU, Federal University of Santa Catarina, Florianópolis, Brazil
| | - J C Nunes
- Pathology Division, HU, Federal University of Santa Catarina, Florianópolis, Brazil
| | - J Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA,Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Z A Bortolotto
- Laboratory of Neurosciences, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, Brazil,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - H J Markowitsch
- Physiological Psychology, University of Bielefeld, Bielefeld, Germany
| | - S L Lightman
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK,Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK
| | - R Walz
- Center for Applied Neuroscience, Hospital Universitário, Federal University of Santa Catarina, Florianópolis, Brazil,Epilepsy Center of Santa Catarina, Federal University of Santa Catarina, Florianópolis, Brazil,Department of Internal Medicine, Federal University of Santa Catarina, Florianópolis, Brazil,Departamento de Clínica Médica, 3 andar, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis CEP 88.040-970, Brazil. E-mail:
| |
Collapse
|