1
|
Guo H, Chen X, Li J, Mo G, Li Y, Tang Y, Kai Y, Zhang S. β-Sitosterol inhibits osteoclast activity and reduces ovariectomy-induced bone loss by regulating the cAMP and NF-κB signaling pathways. Cell Signal 2025; 130:111672. [PMID: 39983806 DOI: 10.1016/j.cellsig.2025.111672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/23/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND β-Sitosterol, a prominent phytosterol present in numerous plant species, has been extensively studied for its potential health benefits, such as lipid-lowering, anxiolytic, and anti-inflammatory properties. Recently, the benefit of β-sitosterol on bone metabolism has been noted. The objective of the current study was to examine the impact of β-sitosterol on the skeletal system. METHODS Network pharmacology and molecular docking were used to predict how β-sitosterol may be used to treat osteoporosis. Cytotoxicity tests were conducted with different concentrations of β-sitosterol. The ability of β-sitosterol to inhibit osteoclast formation and function was evaluated, along with its potential molecular mechanism. An ovariectomized mouse model was used to assess the preventive effect of β-sitosterol on bone loss. RESULTS Network pharmacology analysis suggested that β-sitosterol could be a potential therapeutic treatment for osteoporosis by regulating the cAMP signaling pathway. β-sitosterol dose-dependently inhibited osteoclast differentiation and function without obvious cytotoxicity. Specifically, 20 μM β-sitosterol could obviously repress the number and size of osteoclasts, decrease the formation of F-actin belts, and reduce the bone-resorbing activity of osteoclasts. Some key signaling mediators, including PKA, c-Jun, NFATc1, p-CREB, and NF-κB, were downregulated by β-sitosterol. β-sitosterol acted by attenuating the cAMP and NF-κB signaling pathways. In vivo experiments confirmed β-sitosterol protected ovariectomy-induced bone loss though suppressing osteoclastic bone resorption. CONCLUSION β-sitosterol could inhibit the production and function of osteoclasts in vitro and reverse ovariectomy-induced bone loss. Thus, β-sitosterol could be a potential supplement for diseases with active bone resorption such as osteoporosis.
Collapse
Affiliation(s)
- Huizhi Guo
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojun Chen
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Jinglan Li
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoye Mo
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongxian Li
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongchao Tang
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Kai
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuncong Zhang
- Spine Surgery Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
2
|
Li M, Li G, Liu Y, Li J, Ou Y, Guan W, Zeng Z, Tang H, Bai D, Zhang G, Huang P, Song L, Ye L, Ke H, Yao H. Design, Synthesis, and Evaluation of Selective PDE4 Inhibitors for the Therapy of Pulmonary Injury. J Med Chem 2025; 68:3837-3857. [PMID: 39847693 DOI: 10.1021/acs.jmedchem.4c02969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Pulmonary inflammation is the main cause of lung injury. Phosphodiesterase 4 (PDE4) is a promising anti-inflammatory target for the treatment of respiratory diseases. Herein, we designed and synthesized 43 compounds in two novel series of benzimidazole derivatives as PDE4 inhibitors. Among them, compound A5 showed highly selective inhibition of PDE4, good safety, and liver microsomal stability in vitro. A5 administration remarkably attenuated inflammatory infiltration and pathologic injury of the lung in models of acute lung injury in mice and chronic obstructive pulmonary disease (COPD) in mice. In addition, A5 enhanced sputum secretion, relieved cough in mice, and inhibited phosphorylation of p38 MAP kinase, an important protein in the regulation of lung injury. Overall, A5, as an effective PDE4 inhibitor without acute toxicity and gastrointestinal reaction, may be a potent candidate for the treatment of pulmonary injury.
Collapse
Affiliation(s)
- Mengjie Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Gang Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Yuanhui Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangdong 510006, China
| | - Jiayu Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Yanghui Ou
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Wen Guan
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Zhijun Zeng
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Dan Bai
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Guoping Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Peiming Huang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Liyan Song
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| | - Lianbao Ye
- School of Pharmacy, Guangdong Pharmaceutical University, Guangdong 510006, China
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, 120 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Hongliang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510260, China
| |
Collapse
|
3
|
Lee E, Chang Y. Modulating Neuroinflammation as a Prospective Therapeutic Target in Alzheimer's Disease. Cells 2025; 14:168. [PMID: 39936960 PMCID: PMC11817173 DOI: 10.3390/cells14030168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
The recent approval of lecanemab highlights that the amyloid beta (Aβ) protein is an important pathological target in Alzheimer's disease (AD) and further emphasizes the significance of neuroinflammatory pathways in regulating Aβ accumulation. Indeed, Aβ accumulation triggers microglia activation, which are key mediators in neuroinflammation. The inflammatory responses in this process can lead to neuronal damage and functional decline. Microglia secrete proinflammatory cytokines that accelerate neuronal death and release anti-inflammatory cytokines and growth factors contributing to neuronal recovery and protection. Thus, microglia play a dual role in neurodegeneration and neuroprotection, complicating their function in AD. Therefore, elucidating the complex interactions between Aβ protein, microglia, and neuroinflammation is essential for developing new strategies for treating AD. This review investigates the receptors and pathways involved in activating microglia and aims to enhance understanding of how these processes impact neuroinflammation in AD, as well as how they can be regulated. This review also analyzed studies reported in the existing literature and ongoing clinical trials. Overall, these studies will contribute to understanding the regulatory mechanisms of neuroinflammation and developing new therapies that can slow the pathological progression of AD.
Collapse
Affiliation(s)
- Eunshil Lee
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Yongmin Chang
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Radiology, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
4
|
Donders Z, Skorupska IJ, Willems E, Mussen F, Broeckhoven JV, Carlier A, Schepers M, Vanmierlo T. Beyond PDE4 inhibition: A comprehensive review on downstream cAMP signaling in the central nervous system. Biomed Pharmacother 2024; 177:117009. [PMID: 38908196 DOI: 10.1016/j.biopha.2024.117009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates signal transduction pathways pivotal for numerous biological functions. Intracellular cAMP levels are spatiotemporally regulated by their hydrolyzing enzymes called phosphodiesterases (PDEs). It has been shown that increased cAMP levels in the central nervous system (CNS) promote neuroplasticity, neurotransmission, neuronal survival, and myelination while suppressing neuroinflammation. Thus, elevating cAMP levels through PDE inhibition provides a therapeutic approach for multiple CNS disorders, including multiple sclerosis, stroke, spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, and Alzheimer's disease. In particular, inhibition of the cAMP-specific PDE4 subfamily is widely studied because of its high expression in the CNS. So far, the clinical translation of full PDE4 inhibitors has been hampered because of dose-limiting side effects. Hence, focusing on signaling cascades downstream activated upon PDE4 inhibition presents a promising strategy, offering novel and pharmacologically safe targets for treating CNS disorders. Yet, the underlying downstream signaling pathways activated upon PDE(4) inhibition remain partially elusive. This review provides a comprehensive overview of the existing knowledge regarding downstream mediators of cAMP signaling induced by PDE4 inhibition or cAMP stimulators. Furthermore, we highlight existing gaps and future perspectives that may incentivize additional downstream research concerning PDE(4) inhibition, thereby providing novel therapeutic approaches for CNS disorders.
Collapse
Affiliation(s)
- Zoë Donders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Iga Joanna Skorupska
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Emily Willems
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Femke Mussen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
5
|
Bozkurt A, Karakoy Z, Aydin P, Ozdemir B, Toktay E, Halici Z, Cadirci E. Targeting Aquaporin-5 by Phosphodiesterase 4 Inhibition Offers New Therapeutic Opportunities for Ovarian Ischemia Reperfusion Injury in Rats. Reprod Sci 2024; 31:2021-2031. [PMID: 38453769 PMCID: PMC11217128 DOI: 10.1007/s43032-024-01496-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024]
Abstract
This study aimed to examine the effect of Phosphodiesterase 4 (PDE4) inhibition on Aquaporin-5 (AQP5) and its potential cell signaling pathway in the ovarian ischemia reperfusion (OIR) model. Thirty adult female rats were divided into five groups: Group 1; Control: Sham operation, Group 2; OIR that 3 hour ischemia followed by 3 hour reperfusion, Group 3; OIR + Rolipram 1 mg/kg, Group 4; OIR + Rolipram 3 mg/kg, Group 5; OIR + Rolipram 5 mg/kg. Rolipram was administered intraperitoneally to the rats in groups 3-4 and 5 at determined doses 30 minutes before reperfusion. From ovary tissue; Tumor necrosis factor-a (TNF-α), Cyclic adenosine monophosphate (cAMP), Nuclear factor kappa (NF-κB), Interleukin-6 (IL-6), Phosphodiesterase 4D (PDE4D), Mitogen-activated protein kinase (MAPK) and AQP5 levels were measured by ELISA. We also measured the level of AQP5 in ovary tissue by real-time reverse-transcription polymerase chain reaction (RT-PCR). In the OIR groups; TNF-α, NF-κB, IL-6, MAPK inflammatory levels increased, and cAMP and AQP5 levels decreased, which improved with the administration of rolipram doses. Also histopathological results showed damaged ovarian tissue after OIR, while rolipram administration decrased tissue damage in a dose dependent manner. We propose that the protective effect of PDE4 inhibition in OIR may be regulated by AQP5 and its potential cell signaling pathway and may be a new target in OIR therapy. However, clinical studies are needed to appraise these data in humans.
Collapse
Affiliation(s)
- Ayse Bozkurt
- Faculty of Pharmacy, Department of Pharmacology, Van Yuzuncu Yil University, Van, Turkey
| | - Zeynep Karakoy
- Faculty of Pharmacy, Department of Pharmacology, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Pelin Aydin
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, 25240, Turkey
- Department of Anesthesiology and Reanimation, Educational and Research Hospital, Erzurum, Turkey
| | - Bengul Ozdemir
- Faculty of Medicine, Department of Histology and Embryology, Kafkas University, Kars, Turkey
| | - Erdem Toktay
- Faculty of Medicine, Department of Histology and Embryology, Kafkas University, Kars, Turkey
| | - Zekai Halici
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, 25240, Turkey
- Clinical Research, Development and Design Application and Research Center, Ataturk University, 25240, Erzurum, Turkey
| | - Elif Cadirci
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, 25240, Turkey.
- Clinical Research, Development and Design Application and Research Center, Ataturk University, 25240, Erzurum, Turkey.
| |
Collapse
|
6
|
Delhaye S, Jarjat M, Boulksibat A, Sanchez C, Tempio A, Turtoi A, Giorgi M, Lacas-Gervais S, Baj G, Rovere C, Trezza V, Pellegrini M, Maurin T, Lalli E, Bardoni B. Defects in AMPAR trafficking and microglia activation underlie socio-cognitive deficits associated to decreased expression of phosphodiesterase 2 a. Neurobiol Dis 2024; 191:106393. [PMID: 38154608 DOI: 10.1016/j.nbd.2023.106393] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Phosphodiesterase 2 A (PDE2A) is an enzyme involved in the homeostasis of cAMP and cGMP and is the most highly expressed PDE in human brain regions critical for socio-cognitive behavior. In cerebral cortex and hippocampus, PDE2A expression level is upregulated in Fmr1-KO mice, a model of the Fragile X Syndrome (FXS), the most common form of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Indeed, PDE2A translation is negatively modulated by FMRP, whose functional absence causes FXS. While the pharmacological inhibition of PDE2A has been associated to its pro-cognitive role in normal animals and in models of ID and ASD, homozygous PDE2A mutations have been identified in patients affected by ID, ASD and epilepsy. To clarify this apparent paradox about the role of PDE2A in brain development, we characterized here Pde2a+/- mice (homozygote animals being not viable) at the behavioral, cellular, molecular and electrophysiological levels. Pde2a+/- females display a milder form of the disorder with reduced cognitive performance in adulthood, conversely males show severe socio-cognitive deficits throughout their life. In males, these phenotypes are associated with microglia activation, elevated glutathione levels and increased externalization of Glutamate receptor (GluR1) in CA1, producing reduced mGluR-dependent Long-term Depression. Overall, our results reveal molecular targets of the PDE2A-dependent pathway underlying socio-cognitive performance. These results clarify the mechanism of action of pro-cognitive drugs based on PDE2A inactivation, which have been shown to be promising therapeutic approaches for Alzheimer's disease, schizophrenia, FXS as well as other forms of ASD.
Collapse
Affiliation(s)
- Sébastien Delhaye
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Marielle Jarjat
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Asma Boulksibat
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Clara Sanchez
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Alessandra Tempio
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Andrei Turtoi
- Inserm U1194, Université Montpellier, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier Cedex 5, France
| | - Mauro Giorgi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, DAHFMO, Sapienza University of Rome, 00161 Rome, Italy
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée, 06100 Nice, France
| | - Gabriele Baj
- Department of Life Science, University of Trieste, 34100 Trieste, Italy
| | - Carole Rovere
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | | | - Manuela Pellegrini
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, DAHFMO, Sapienza University of Rome, 00161 Rome, Italy; Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015 Monterotondo Scalo, Rome, Italy
| | - Thomas Maurin
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Enzo Lalli
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Barbara Bardoni
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France.
| |
Collapse
|
7
|
Baig TA, Haniffa HM, Siddiqui H, Shah SF, Jabeen A. A new acyl derivative of sulfadimethoxine inhibits phagocyte oxidative burst and ameliorates inflammation in a mice model of zymosan-induced generalised inflammation. Inflammopharmacology 2023; 31:3303-3316. [PMID: 37971604 DOI: 10.1007/s10787-023-01372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/09/2023] [Indexed: 11/19/2023]
Abstract
Chronic inflammation and oxidative stress play a pivotal role in the pathophysiology of most challenging illnesses, including cancer, Alzheimer's, cardiovascular and autoimmune diseases. The present study aimed to investigate the anti-inflammatory potential of a new sulfadimethoxine derivative N-(4-(N-(2,6-dimethoxypyrimidin-4-yl) sulfamoyl) phenyl) dodecanamide (MHH-II-32). The compound was characterised by applying 1H-, 13C-NMR, EI-MS and HRFAB-MS spectroscopic techniques. The compound inhibited zymosan-induced oxidative bursts from whole blood phagocytes and isolated polymorphonuclear cells with an IC50 value of (2.5 ± 0.4 and 3.4 ± 0.3 µg/mL), respectively. Furthermore, the inhibition of nitric oxide with an IC50 (3.6 ± 2.2 µg/mL) from lipopolysaccharide-induced J774.2 macrophages indicates its in vitro anti-inflammatory efficacy. The compound did not show toxicity towards normal fibroblast cells. The observational findings, gross anatomical analysis of visceral organs and serological tests revealed the non-toxicity of the compound at the highest tested intraperitoneal (IP) dose of 100 mg/kg in acute toxicological studies in Balb/c mice. The compound treatment (100 mg/kg) (SC) significantly (P < 0.001) downregulated the mRNA expression of inflammatory markers TNF-α, IL-1β, IL-2, IL-13, and NF-κB, which were elevated in zymosan-induced generalised inflammation (IP) in Balb/c mice while upregulated the expression of anti-inflammatory cytokine IL-10, which was reduced in zymosan-treated mice. No suppressive effect was observed at the dose of 25 mg/kg. Ibuprofen was taken as a standard drug. The results revealed that the new acyl derivative of sulfadimethoxine has an immunomodulatory effect against generalised inflammatory response with non-toxicity both in vitro and in vivo, and has therapeutic potential for various chronic inflammatory illnesses.
Collapse
Affiliation(s)
- Tariq Ahmad Baig
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Haroon M Haniffa
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Department of Chemical Sciences, Faculty of Applied Sciences, South Eastern University, Sammanthurai, 32200, Sri Lanka
| | - Hina Siddiqui
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Syeda Farah Shah
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Almas Jabeen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
8
|
Lummertz Magenis M, Souza de Marcos P, Paganini Damiani A, Ricardo Cantareli da Silva A, Martins Longaretti L, Bahia Franca I, Da Silva J, Rodrigues Boeck C, Moraes de Andrade V. Genotoxic effects of caffeine in female mice exposed during pregnancy and lactation period and their offspring. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2023; 41:36-60. [PMID: 37243358 DOI: 10.1080/26896583.2023.2213613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Caffeine is a widely consumed substance, and there is a discussion about its effects when ingested by women during pregnancy and lactation. We aimed to identify the genotoxic effects of caffeine in female mice that consumed it during pregnancy and lactation periods and its consequences in their offspring. Thirty-six couples of Swiss mice received water or caffeine (0.3 and 1.0 mg/mL) treatment during pregnancy and lactation. The male and female offspring were divided into 12 groups according to the treatment administered to the female mice. Genotoxicity was assessed using the comet assay and the micronucleus test. Both doses of caffeine showed genotoxic effects in pregnant and lactating mice groups compared to groups not administered caffeine. In relation to offspring, it can be observed that females and males of the offspring had low weight in early life. In both female and male offspring, genotoxicity was detected in the blood, liver, and kidney tissues. Thus, from the present study, we can suggest that the caffeine consumed by female mice during the periods of pregnancy and lactation led to genotoxic effects in their offspring.
Collapse
Affiliation(s)
- Marina Lummertz Magenis
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Pamela Souza de Marcos
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Adriani Paganini Damiani
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Anderson Ricardo Cantareli da Silva
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Luiza Martins Longaretti
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Ive Bahia Franca
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - Juliana Da Silva
- Laboratory of Genetic Toxicology, Lutheran University of Brazil, Canoas, Rio Grande do Sul, Brazil
- La Salle University, Canoas, Rio Grande do Sul, Brazil
| | - Carina Rodrigues Boeck
- Graduate Program in Nanosciences, Master Degree in Health and Life Science, Franciscan University, Santa Maria, Rio Grande do Sul, Brazil
| | - Vanessa Moraes de Andrade
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina, Criciúma, Santa Catarina, Brazil
| |
Collapse
|
9
|
Mussen F, Broeckhoven JV, Hellings N, Schepers M, Vanmierlo T. Unleashing Spinal Cord Repair: The Role of cAMP-Specific PDE Inhibition in Attenuating Neuroinflammation and Boosting Regeneration after Traumatic Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24098135. [PMID: 37175842 PMCID: PMC10179671 DOI: 10.3390/ijms24098135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is characterized by severe neuroinflammation and hampered neuroregeneration, which often leads to permanent neurological deficits. Current therapies include decompression surgery, rehabilitation, and in some instances, the use of corticosteroids. However, the golden standard of corticosteroids still achieves minimal improvements in functional outcomes. Therefore, new strategies tackling the initial inflammatory reactions and stimulating endogenous repair in later stages are crucial to achieving functional repair in SCI patients. Cyclic adenosine monophosphate (cAMP) is an important second messenger in the central nervous system (CNS) that modulates these processes. A sustained drop in cAMP levels is observed during SCI, and elevating cAMP is associated with improved functional outcomes in experimental models. cAMP is regulated in a spatiotemporal manner by its hydrolyzing enzyme phosphodiesterase (PDE). Growing evidence suggests that inhibition of cAMP-specific PDEs (PDE4, PDE7, and PDE8) is an important strategy to orchestrate neuroinflammation and regeneration in the CNS. Therefore, this review focuses on the current evidence related to the immunomodulatory and neuroregenerative role of cAMP-specific PDE inhibition in the SCI pathophysiology.
Collapse
Affiliation(s)
- Femke Mussen
- Department of Neuroscience, Biomedical Research Institute BIOMED, Hasselt University, 3590 Diepenbeek, Belgium
- University MS Center (UMSC) Hasselt-Pelt, Hasselt University, 3500 Hasselt, Belgium
| | - Jana Van Broeckhoven
- University MS Center (UMSC) Hasselt-Pelt, Hasselt University, 3500 Hasselt, Belgium
- Department of Immunology and Infection, Biomedical Research Institute BIOMED, Hasselt University, 3590 Diepenbeek, Belgium
| | - Niels Hellings
- University MS Center (UMSC) Hasselt-Pelt, Hasselt University, 3500 Hasselt, Belgium
- Department of Immunology and Infection, Biomedical Research Institute BIOMED, Hasselt University, 3590 Diepenbeek, Belgium
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute BIOMED, Hasselt University, 3590 Diepenbeek, Belgium
- University MS Center (UMSC) Hasselt-Pelt, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, The Netherlands
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute BIOMED, Hasselt University, 3590 Diepenbeek, Belgium
- University MS Center (UMSC) Hasselt-Pelt, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, The Netherlands
| |
Collapse
|
10
|
Yu M, Ou Y, Wang H, Gu W. PU.1 interaction with p50 promotes microglial-mediated inflammation in secondary spinal cord injury in SCI rats. Int J Neurosci 2023; 133:389-402. [PMID: 33970748 DOI: 10.1080/00207454.2021.1923017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose/aim of the study Secondary spinal cord injury is the inflammatory damage to surrounding tissues caused by activated microglial-mediated neuroinflammatory responses. The nuclear factor-κB (p65/p50) pathway and PU.1 are closely correlated with inflammatory responses; thus, we examined the relationship and function between PU.1 and p50 in secondary spinal cord injury.Materials and methods In this study, we established an adult rat acute spinal cord injury model to simulate the pathological process of spinal cord injury.Results: We found that the expression of PU.1 was significantly increased at three days after spinal cord injury and mainly expressed in activated microglia. Moreover, p-p50 expression was increased in SCI rats and the protein interacted with PU.1. Lipopolysaccharide was used to induce microglia activation in vitro.Conclusions: The results showed that PU.1 and p-p50 expression was significantly increased and PU.1 interacted with p50 in the nucleus. The levels of tumor necrosis factor-α and interleukin-1β secreted by microglia were detected by enzyme-linked immunosorbent assay. The results showed that when both PU.1 and p50 were overexpressed, tumor necrosis factor-α and interleukin-1β secretion was significantly increased to levels higher than in cells overexpressing PU.1 or p50 alone. These results suggest that PU.1 and p50 interact to promote p65 transcription and the expression of inflammatory factors, which is an important mechanism of the microglial-mediated inflammatory response to secondary injury after spinal cord injury.
Collapse
Affiliation(s)
- Mingchen Yu
- Department of Orthopedics, Changzhou Seventh People's Hospital, Changzhou, Jiangsu Province, China.,Nantong University, Nantong, Jiangsu Province, China
| | - Yiqing Ou
- Nantong University, Nantong, Jiangsu Province, China.,The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Hongmei Wang
- Nantong University, Nantong, Jiangsu Province, China
| | - Weidong Gu
- Department of Orthopedics, Changzhou Seventh People's Hospital, Changzhou, Jiangsu Province, China
| |
Collapse
|
11
|
Anti-Inflammatory and Neuroprotective Effects of Morin in an MPTP-Induced Parkinson’s Disease Model. Int J Mol Sci 2022; 23:ijms231810578. [PMID: 36142491 PMCID: PMC9501291 DOI: 10.3390/ijms231810578] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022] Open
Abstract
Neurodegenerative diseases such as Parkinson’s disease (PD) are known to be related to oxidative stress and neuroinflammation, and thus, modulating neuroinflammation offers a possible means of treating PD-associated pathologies. Morin (2′,3,4′,5,7-pentahydroxy flavone) is a flavonol with anti-oxidative and anti-inflammatory effects found in wines, herbs, and fruits. The present study was undertaken to determine whether a morin-containing diet has protective effects in an MPTP-induced mouse model of PD. Mice were fed a control or morin diet for 34 days, and then MPTP (30 mg/kg, i.p.) was administered daily for 5 days to induce a PD-like pathology. We found that dietary morin prevented MPTP-induced motor dysfunction and ameliorated dopaminergic neuronal damage in striatum (STR) and substantia nigra (SN) in our mouse model. Furthermore, MPTP-induced neuroinflammation was significantly reduced in mice fed morin. In vitro studies showed that morin effectively suppressed glial activations in primary microglia and astrocytes, and biochemical analysis and a docking simulation indicated that the anti-inflammatory effects of morin were mediated by blocking the extracellular signal-regulated kinase (ERK)-p65 pathway. These findings suggest that morin effectively inhibits glial activations and has potential use as a functional food ingredient with therapeutic potential for the treatment of PD and other neurodegenerative diseases associated with neuroinflammation.
Collapse
|
12
|
Li R, Liang Y, Lin B. Accumulation of systematic TPM1 mediates inflammation and neuronal remodeling by phosphorylating PKA and regulating the FABP5/NF-κB signaling pathway in the retina of aged mice. Aging Cell 2022; 21:e13566. [PMID: 35148456 PMCID: PMC8920455 DOI: 10.1111/acel.13566] [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: 09/08/2021] [Revised: 01/05/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
The molecular mechanisms underlying functional decline during normal brain aging are poorly understood. Here, we identified the actin‐associated protein tropomyosin 1 (TPM1) as a new systemic pro‐aging factor associated with function deficits in normal aging retinas. Heterochronic parabiosis and blood plasma treatment confirmed that systemic factors regulated age‐related inflammatory responses and the ectopic dendritic sprouting of rod bipolar (RBC) and horizontal (HC) cells in the aging retina. Proteomic analysis revealed that TPM1 was a potential systemic molecule underlying structural and functional deficits in the aging retina. Recombinant TPM1 protein administration accelerated the activation of glial cells, the dendritic sprouting of RBCs and HCs and functional decline in the retina of young mice, whereas anti‐TPM1 neutralizing antibody treatment ameliorated age‐related structural and function changes in the retina of aged mice. Old mouse plasma (OMP) induced glial cell activation and the dendritic outgrowth of RBCs and HCs in young mice, and yet TMP1‐depleted OMP failed to reproduce the similar effect in young mice. These results confirmed that TPM1 was a systemic pro‐aging factor. Moreover, we demonstrated that systematic TPM1 was an immune‐related molecule, which elicited endogenous TPM1 expression and inflammation by phosphorylating PKA and regulating FABP5/NF‐κB signaling pathway in normal aging retinas. Interestingly, we observed TPM1 upregulation and the ectopic dendritic sprouting of RBCs and HCs in young mouse models of Alzheimer's disease, indicating a potential role of TPM1 in age‐related neurodegenerative diseases. Our data indicate that TPM1 could be targeted for combating the aging process.
Collapse
Affiliation(s)
- Rong Li
- School of Optometry The Hong Kong Polytechnic University Kowloon Hong Kong
| | - Yuxiang Liang
- The State Key Laboratory of Brain and Cognitive Sciences The University of Hong Kong Pok Fu Lam Hong Kong
| | - Bin Lin
- School of Optometry The Hong Kong Polytechnic University Kowloon Hong Kong
| |
Collapse
|
13
|
Zhou G, Wang Z, Han S, Chen X, Li Z, Hu X, Li Y, Gao J. Multifaceted Roles of cAMP Signaling in the Repair Process of Spinal Cord Injury and Related Combination Treatments. Front Mol Neurosci 2022; 15:808510. [PMID: 35283731 PMCID: PMC8904388 DOI: 10.3389/fnmol.2022.808510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/26/2022] [Indexed: 01/03/2023] Open
Abstract
Spinal cord injury (SCI) results in multiple pathophysiological processes, including blood–spinal cord barrier disruption, hemorrhage/ischemia, oxidative stress, neuroinflammation, scar formation, and demyelination. These responses eventually lead to severe tissue destruction and an inhibitory environment for neural regeneration.cAMP signaling is vital for neurite outgrowth and axonal guidance. Stimulating intracellular cAMP activity significantly promotes neuronal survival and axonal regrowth after SCI.However, neuronal cAMP levels in adult CNS are relatively low and will further decrease after injury. Targeting cAMP signaling has become a promising strategy for neural regeneration over the past two decades. Furthermore, studies have revealed that cAMP signaling is involved in the regulation of glial cell function in the microenvironment of SCI, including macrophages/microglia, reactive astrocytes, and oligodendrocytes. cAMP-elevating agents in the post-injury milieu increase the cAMP levels in both neurons and glial cells and facilitate injury repair through the interplay between neurons and glial cells and ultimately contribute to better morphological and functional outcomes. In recent years, combination treatments associated with cAMP signaling have been shown to exert synergistic effects on the recovery of SCI. Agents carried by nanoparticles exhibit increased water solubility and capacity to cross the blood–spinal cord barrier. Implanted bioscaffolds and injected hydrogels are potential carriers to release agents locally to avoid systemic side effects. Cell transplantation may provide permissive matrices to synergize with the cAMP-enhanced growth capacity of neurons. cAMP can also induce the oriented differentiation of transplanted neural stem/progenitor cells into neurons and increase the survival rate of cell grafts. Emerging progress focused on cAMP compartmentation provides researchers with new perspectives to understand the complexity of downstream signaling, which may facilitate the clinical translation of strategies targeting cAMP signaling for SCI repair.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhiyan Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shiyuan Han
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhimin Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianghui Hu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of International Medical Service, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Jun Gao
| |
Collapse
|
14
|
Markitantova YV, Simirskii VN. The Role of the Purinergic Signaling System in the Control of Histogenesis, Homeostasis, and Pathogenesis of the Vertebrate Retina. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
Joshi S, Gomes ED, Wang T, Corben A, Taldone T, Gandu S, Xu C, Sharma S, Buddaseth S, Yan P, Chan LYL, Gokce A, Rajasekhar VK, Shrestha L, Panchal P, Almodovar J, Digwal CS, Rodina A, Merugu S, Pillarsetty N, Miclea V, Peter RI, Wang W, Ginsberg SD, Tang L, Mattar M, de Stanchina E, Yu KH, Lowery M, Grbovic-Huezo O, O'Reilly EM, Janjigian Y, Healey JH, Jarnagin WR, Allen PJ, Sander C, Erdjument-Bromage H, Neubert TA, Leach SD, Chiosis G. Pharmacologically controlling protein-protein interactions through epichaperomes for therapeutic vulnerability in cancer. Commun Biol 2021; 4:1333. [PMID: 34824367 PMCID: PMC8617294 DOI: 10.1038/s42003-021-02842-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 11/03/2021] [Indexed: 12/03/2022] Open
Abstract
Cancer cell plasticity due to the dynamic architecture of interactome networks provides a vexing outlet for therapy evasion. Here, through chemical biology approaches for systems level exploration of protein connectivity changes applied to pancreatic cancer cell lines, patient biospecimens, and cell- and patient-derived xenografts in mice, we demonstrate interactomes can be re-engineered for vulnerability. By manipulating epichaperomes pharmacologically, we control and anticipate how thousands of proteins interact in real-time within tumours. Further, we can essentially force tumours into interactome hyperconnectivity and maximal protein-protein interaction capacity, a state whereby no rebound pathways can be deployed and where alternative signalling is supressed. This approach therefore primes interactomes to enhance vulnerability and improve treatment efficacy, enabling therapeutics with traditionally poor performance to become highly efficacious. These findings provide proof-of-principle for a paradigm to overcome drug resistance through pharmacologic manipulation of proteome-wide protein-protein interaction networks.
Collapse
Affiliation(s)
- Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Erica DaGama Gomes
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Adriana Corben
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Tony Taldone
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Srinivasa Gandu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Xu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Salma Buddaseth
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lon Yin L Chan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Askan Gokce
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Vinagolu K Rajasekhar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lisa Shrestha
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Justina Almodovar
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Swathi Merugu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Vlad Miclea
- Faculty of Automation and Computer Science, Technical University of Cluj-Napoca, Cluj-Napoca, CJ, 400114, Romania
| | - Radu I Peter
- Faculty of Automation and Computer Science, Technical University of Cluj-Napoca, Cluj-Napoca, CJ, 400114, Romania
| | - Wanyan Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Neuroscience & Physiology, and the NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Laura Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Marissa Mattar
- Antitumour Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Elisa de Stanchina
- Antitumour Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Kenneth H Yu
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Maeve Lowery
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Olivera Grbovic-Huezo
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Eileen M O'Reilly
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yelena Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, 10065, USA
| | - John H Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - William R Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Peter J Allen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Chris Sander
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, NYU School of Medicine, New York, NY, 10016, USA
| | - Steven D Leach
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Dartmouth Geisel School of Medicine and Norris Cotton Cancer Center, Lebanon, NH, 03766, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
16
|
Carregosa D, Mota S, Ferreira S, Alves-Dias B, Loncarevic-Vasiljkovic N, Crespo CL, Menezes R, Teodoro R, dos Santos CN. Overview of Beneficial Effects of (Poly)phenol Metabolites in the Context of Neurodegenerative Diseases on Model Organisms. Nutrients 2021; 13:2940. [PMID: 34578818 PMCID: PMC8464690 DOI: 10.3390/nu13092940] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
The rise of neurodegenerative diseases in an aging population is an increasing problem of health, social and economic consequences. Epidemiological and intervention studies have demonstrated that diets rich in (poly)phenols can have potent health benefits on cognitive decline and neurodegenerative diseases. Meanwhile, the role of gut microbiota is ever more evident in modulating the catabolism of (poly)phenols to dozens of low molecular weight (poly)phenol metabolites that have been identified in plasma and urine. These metabolites can reach circulation in higher concentrations than parent (poly)phenols and persist for longer periods of time. However, studies addressing their potential brain effects are still lacking. In this review, we will discuss different model organisms that have been used to study how low molecular weight (poly)phenol metabolites affect neuronal related mechanisms gathering critical insight on their potential to tackle the major hallmarks of neurodegeneration.
Collapse
Affiliation(s)
- Diogo Carregosa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Sara Mota
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- iBET, Institute of Experimental and Technological Biology, Apartado 12, 2781-901 Oeiras, Portugal
| | - Sofia Ferreira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- CBIOS, University Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Beatriz Alves-Dias
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Natasa Loncarevic-Vasiljkovic
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Carolina Lage Crespo
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Regina Menezes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- iBET, Institute of Experimental and Technological Biology, Apartado 12, 2781-901 Oeiras, Portugal
- CBIOS, University Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Rita Teodoro
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Cláudia Nunes dos Santos
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- iBET, Institute of Experimental and Technological Biology, Apartado 12, 2781-901 Oeiras, Portugal
| |
Collapse
|
17
|
Morris G, Walder K, Kloiber S, Amminger P, Berk M, Bortolasci CC, Maes M, Puri BK, Carvalho AF. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res 2021; 170:105729. [PMID: 34119623 DOI: 10.1016/j.phrs.2021.105729] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The endocannabinoid system (ECS) comprises two cognate endocannabinoid receptors referred to as CB1R and CB2R. ECS dysregulation is apparent in neurodegenerative/neuro-psychiatric disorders including but not limited to schizophrenia, major depressive disorder and potentially bipolar disorder. The aim of this paper is to review mechanisms whereby both receptors may interact with neuro-immune and neuro-oxidative pathways, which play a pathophysiological role in these disorders. CB1R is located in the presynaptic terminals of GABAergic, glutamatergic, cholinergic, noradrenergic and serotonergic neurons where it regulates the retrograde suppression of neurotransmission. CB1R plays a key role in long-term depression, and, to a lesser extent, long-term potentiation, thereby modulating synaptic transmission and mediating learning and memory. Optimal CB1R activity plays an essential neuroprotective role by providing a defense against the development of glutamate-mediated excitotoxicity, which is achieved, at least in part, by impeding AMPA-mediated increase in intracellular calcium overload and oxidative stress. Moreover, CB1R activity enables optimal neuron-glial communication and the function of the neurovascular unit. CB2R receptors are detected in peripheral immune cells and also in central nervous system regions including the striatum, basal ganglia, frontal cortex, hippocampus, amygdala as well as the ventral tegmental area. CB2R upregulation inhibits the presynaptic release of glutamate in several brain regions. CB2R activation also decreases neuroinflammation partly by mediating the transition from a predominantly neurotoxic "M1" microglial phenotype to a more neuroprotective "M2" phenotype. CB1R and CB2R are thus novel drug targets for the treatment of neuro-immune and neuro-oxidative disorders including schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| |
Collapse
|
18
|
Muñoz-Pérez VM, Ortiz MI, Salas-Casa A, Pérez-Guerrero J, Castillo-Pacheco N, Barragán-Ramírez G, Hernándes-Alejandro M. In vitro effects of citral on the human myometrium: Potential adjunct therapy to prevent preterm births. Birth Defects Res 2021; 113:613-622. [PMID: 33484091 DOI: 10.1002/bdr2.1873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Premature infants contribute to infant morbidity and mortality especially in low resource settings. Information on tocolytic and/or anti-inflammatory effects of several plant extracts, such as citral, could help prevent preterm birth cases and reduce the number of preterm infants. The aim of this study was to evaluate the in vitro tocolytic and anti-inflammatory effect of citral on myometrial tissues of the human uterus. METHODS Myometrial samples from uteri obtained after hysterectomy were used in functional tests to evaluate the inhibitory effect of citral on PGF-2α induced contractions. The intracellular cyclic adenosine monophosphate (cAMP) levels generated in response to citral in human myometrial homogenates were measured by ELISAs. Forskolin was used as a positive control. The anti-inflammatory effect of citral was determined through the measurement of two pro-inflammatory cytokines, tumor necrosis factor-α (TNFα) and interleukin (IL)-1β, and the anti-inflammatory cytokine IL-10, in human myometrial explants stimulated with lipopolysaccharide (LPS). RESULTS Citral was able to induce a significant inhibition of PGF-2α induced contractions at the highest concentration level (p < .05). Citral caused a concentration-dependent increase in myometrial cAMP levels (p < .05) and a concentration-dependent decrease in LPS-induced TNFα and IL-1β production, while IL-10 production increased significantly (p < .05). The anti-inflammatory and tocolytic effects induced by citral could be associated with an increase in cAMP levels in human myometrial samples. CONCLUSION These properties place citral as a potentially safe and effective adjuvant agent in preterm birth cases, an obstetric and gynecological problem that requires urgent attention.
Collapse
Affiliation(s)
- Víctor Manuel Muñoz-Pérez
- Área Académica de Medicina del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Mario I Ortiz
- Área Académica de Medicina del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Andrés Salas-Casa
- Área Académica de Gerontología del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Mexico
| | - Jessica Pérez-Guerrero
- Departamento de Ginecología y Obstetricia del Hospital General de los SSH, Pachuca, Mexico
| | - Narmi Castillo-Pacheco
- Departamento de Ginecología y Obstetricia del Hospital General de los SSH, Pachuca, Mexico
| | | | - Mario Hernándes-Alejandro
- Departamento de Bioingeniería, Unidad profesional Interdisciplinaria de biotecnología del Instituto Politécnico Nacional (UPIBI-IPN), México City, Mexico
| |
Collapse
|
19
|
Kowalska K, Habrowska-Górczyńska DE, Kozieł MJ, Urbanek KA, Domińska K, Piastowska-Ciesielska AW. Mycotoxin Alternariol (AOH) Affects Viability and Motility of Mammary Breast Epithelial Cells. Int J Mol Sci 2021; 22:ijms22020696. [PMID: 33445675 PMCID: PMC7828135 DOI: 10.3390/ijms22020696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/09/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
Mycotoxins are present in everyday diet as common food and feed pollutants. A part of them is still concerned as so-called emerging mycotoxins. Due to the lack of toxicity data, the safety limits and detail molecular mechanism have been not established yet for all of them. Alternariol (AOH), as one of these mycotoxins, produced by Alternaria species, is so far reported as an estrogenic, genotoxic, and immunomodulatory agent; however, its direct effect on human health is not known. Especially, in the case of hormone-dependent tissues which are sensitive to both endogenic, as well as external estrogenic agents, it might be crucial to assess the effect of AOH. Thus, this study evaluated how exposure to AOH affects viability and motility of the human normal mammary gland epithelial in vitro model. We observed that AOH significantly affects viability of cells in a time- and dose-dependent manner. Moreover, the induction of oxidative stress, DNA damage, and cell cycle arrest in the G2/M cell cycle phase was observed. The motility of 184A1 cells was also significantly affected. On the molecular level, AOH induced antioxidative stress response via activation of Nuclear factor erythroid 2-related factor 2 (NRF2) signaling pathway agents, as well as decrease in the phosphorylation of protein kinase B (Akt) and p44/42 (ERK 1-2) molecules, indicating that AOH might affect crucial signaling pathways in both physiological and pathophysiological processes in breast tissue.
Collapse
Affiliation(s)
- Karolina Kowalska
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, Zeligowskiego 7/9, 90-752 Lodz, Poland; (D.E.H.-G.); (M.J.K.); (K.A.U.)
- Correspondence: (K.K.); (A.W.P.-C.)
| | - Dominika Ewa Habrowska-Górczyńska
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, Zeligowskiego 7/9, 90-752 Lodz, Poland; (D.E.H.-G.); (M.J.K.); (K.A.U.)
| | - Marta Justyna Kozieł
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, Zeligowskiego 7/9, 90-752 Lodz, Poland; (D.E.H.-G.); (M.J.K.); (K.A.U.)
| | - Kinga Anna Urbanek
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, Zeligowskiego 7/9, 90-752 Lodz, Poland; (D.E.H.-G.); (M.J.K.); (K.A.U.)
| | - Kamila Domińska
- Medical University of Lodz, Department of Comparative Endocrinology, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | - Agnieszka Wanda Piastowska-Ciesielska
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, Zeligowskiego 7/9, 90-752 Lodz, Poland; (D.E.H.-G.); (M.J.K.); (K.A.U.)
- Correspondence: (K.K.); (A.W.P.-C.)
| |
Collapse
|
20
|
Tao J, Yin S, Song Y, Zeng L, Li S, Liu N, Sun H, Fu Z, Wang Y, Li Y, Liu Y, Sun J, Wang Y, Yang X. Novel scorpion venom peptide HsTx2 ameliorates cerebral ischemic brain injury in rats via the MAPK signaling pathway. Biochem Biophys Res Commun 2020; 534:442-449. [PMID: 33248693 DOI: 10.1016/j.bbrc.2020.11.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/14/2020] [Indexed: 12/18/2022]
Abstract
Ischemic stroke is a severe threat to human health due to its high recurrence, mortality, and disability rates. As such, how to prevent and treat ischemic stroke effectively has become a research hotspot in recent years. Here, we identified a novel peptide, named HsTx2 (AGKKERAGSRRTKIVMLKCIREHGH, 2 861.855 Da), derived from the scorpion Heterometrus spinifer, which showed obvious anti-apoplectic effects in rats with ischemic stroke. Results further demonstrated that HsTx2 significantly reduced formation of infarct area and improved behavioral abnormalities in ischemic stroke rats. These protective effects were likely exerted via activation of the mitogen-activated protein kinase (MAPK) signaling pathway, i.e., up-regulation of phosphorylated ERK1/2 in both rat cerebral cortex and activated microglia (AM); up-regulation of phosphorylated p38 (p-p38) in the cerebral cortex; and inhibition of phosphorylated JNK and p-p38 levels in the AM. In conclusion, this study highlights HsTx2 as a potential neuroprotective agent for stroke.
Collapse
Affiliation(s)
- Jian Tao
- Department of Pharmacology, Medical School, Kunming University, Kunming, 650214, Yunnan, China
| | - Saige Yin
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yongli Song
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Lin Zeng
- Public Technical Service Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Shanshan Li
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Naixin Liu
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Huiling Sun
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhe Fu
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yinglei Wang
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yilin Li
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yixiang Liu
- Key Laboratory of Chemistry in Ethnic Medicine Resource, State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, Yunnan Minzu University, Kunming, Yunnan, 650504, China
| | - Jun Sun
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicine Resource, State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, Yunnan Minzu University, Kunming, Yunnan, 650504, China.
| | - Xinwang Yang
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
21
|
SARS-CoV-2 transcriptome analysis and molecular cataloguing of immunodominant epitopes for multi-epitope based vaccine design. Genomics 2020; 112:5044-5054. [PMID: 32920121 PMCID: PMC7500163 DOI: 10.1016/j.ygeno.2020.09.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/18/2020] [Accepted: 09/08/2020] [Indexed: 01/12/2023]
Abstract
Genomics-led researches are engaged in tracing virus expression pattern, and induced immune responses in human to develop effective vaccine against COVID-19. In this study, targeted expression profiling and differential gene expression analysis of major histocompatibility complexes and innate immune system genes were performed through SARS-CoV-2 infected RNA-seq data of human cell line, and virus transcriptome was generated for T-and B-cell epitope prediction. Docking studies of epitopes with MHC and B-cell receptors were performed to identify potential T-and B-cell epitopes. Transcriptome analysis revealed the specific multiple allele expressions in cell line, genes for elicited induce immune response, and virus gene expression. Proposed T- and B-cell epitopes have high potential to elicit equivalent immune responses caused by SARS-CoV-2 infection which can be useful to provide links between elicited immune response and virus gene expression. This study will facilitate in vitro and in vivo vaccine related research studies in disease control. SARS-CoV-2 transcriptome construction from RNA-seq data of infected human cell-lines. T- and B-cell epitopes from SARS-CoV-2 transcriptome Gene expression profiling of MHC alleles and innate immune system genes
Collapse
|
22
|
Tibbo AJ, Baillie GS. Phosphodiesterase 4B: Master Regulator of Brain Signaling. Cells 2020; 9:cells9051254. [PMID: 32438615 PMCID: PMC7291338 DOI: 10.3390/cells9051254] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Phosphodiesterases (PDEs) are the only superfamily of enzymes that have the ability to break down cyclic nucleotides and, as such, they have a pivotal role in neurological disease and brain development. PDEs have a modular structure that allows targeting of individual isoforms to discrete brain locations and it is often the location of a PDE that shapes its cellular function. Many of the eleven different families of PDEs have been associated with specific diseases. However, we evaluate the evidence, which suggests the activity from a sub-family of the PDE4 family, namely PDE4B, underpins a range of important functions in the brain that positions the PDE4B enzymes as a therapeutic target for a diverse collection of indications, such as, schizophrenia, neuroinflammation, and cognitive function.
Collapse
|
23
|
Cong Z, Li D, Tao Y, Lv X, Zhu X. α 2A -AR antagonism by BRL-44408 maleate attenuates acute lung injury in rats with downregulation of ERK1/2, p38MAPK, and p65 pathway. J Cell Physiol 2020; 235:6905-6914. [PMID: 32003020 DOI: 10.1002/jcp.29586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/13/2020] [Indexed: 12/17/2022]
Abstract
Acute respiratory distress syndrome (ARDS), characterized by acute hypoxic respiratory dysfunction or failure, is a manifestation of multiple organ failure in the lung, and the most common risk factor is sepsis. We previously showed that blocking α2 -adrenoceptor (α2 -AR) could attenuate lung injury induced by endotoxin in rats. α2A -adrenoceptor (α2A -AR), a subtype of α2 -AR plays a key role in inflammatory diseases, but the mechanism remains unknown. Here, we explored the effect of BRL-44408 maleate (BRL), a specific α2A -AR antagonist, on cecal ligation puncture (CLP)-induced ARDS in rats and the underlying mechanism. Preadministration of BRL-44408 maleate significantly alleviated CLP-induced histological injury, macrophage infiltration, inflammatory response, and wet/dry ratio in lung tissue. However, there was no statistical difference in survival rate between the CLP and CLP+BRL groups. Extracellular regulated protein kinase (ERK1/2), p38MAPK, and p65 were activated in the CLP group, and BRL-44408 maleate inhibited the activation of these signal molecules, c-Jun N-terminal kinase (JNK) and protein kinase A (PKA) showed no changes in activation between these two groups. BRL-44408 maleate decreased lipopolysaccharide (LPS)-induced expression of cytokines in NR8383 rat alveolar macrophages and reduced phosphorylation of ERK1/2, p38MAPK, and p65. JNK and PKA were not influenced by LPS. Together, these findings suggest that antagonism of α2A -AR improves CLP-induced acute lung injury and involves the downregulation of ERK1/2, p38MAPK, and p65 pathway independent of the activation of JNK and PKA.
Collapse
Affiliation(s)
- Zhukai Cong
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Dan Li
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yifan Tao
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiangpeng Lv
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Xi Zhu
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
24
|
Zhang L, Zhang J, Jiang X, Yang L, Zhang Q, Wang B, Cui L, Wang X. Hydroxytyrosol Inhibits LPS-Induced Neuroinflammatory Responses via Suppression of TLR-4-Mediated NF-κB P65 Activation and ERK Signaling Pathway. Neuroscience 2019; 426:189-200. [PMID: 31866556 DOI: 10.1016/j.neuroscience.2019.12.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/26/2019] [Accepted: 12/03/2019] [Indexed: 02/08/2023]
Abstract
Neuroinflammation has been implicated in the mechanism underlying the progression of neurodegeneration and infectious neuropathology. Growing evidence suggest that hydroxytyrosol (3,4-dihydroxyphenil-ethanol, HT), one of the main polyphenols presented in extra virgin olive oil (EVOO), has shown potential anti-inflammatory and neuroprotective effects. However, the potential anti-neuroinflammation activity and underlying mechanism of HT remain poorly understood. The present study aimed to investigate the effects of HT on lipopolysaccharide (LPS)-induced inflammation in both in vitro and in vivo models and the associated molecular mechanism. Our results revealed that HT significantly reduced the production of pro-inflammatory mediators in BV2 microglia and primary microglia cells. Phenotypic analysis showed that HT significantly reduced M1 marker CD86 expression and increased M2 marker CD206 expression. In addition, HT significantly decreased the levels of phospho-NF-κB p65 and phospho-extracellular signal-regulated kinase (ERK) in a dose-dependent manner. Moreover, HT suppressed the LPS-induced Toll like receptor 4 (TLR4) in BV2 microglia. In vivo administration of HT following LPS injection significantly reduced some proinflammatory mediator levels and microglia/astrocyte activation in the brain. Together, these results suggest that HT suppressed the LPS-induced neuroinflammatory responses via modulation of microglia M1/M2 polarization and downregulation of TLR-4 mediated NF-κB activation and ERK signaling pathway.
Collapse
Affiliation(s)
- Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China.
| | - Jinlu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Xiaolin Jiang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Qi Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Botao Wang
- Graduate School, Tianjin Medical University, Tianjin 300070, China
| | - Lihua Cui
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Ximo Wang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China.
| |
Collapse
|
25
|
Lee YY, Park JS, Leem YH, Park JE, Kim DY, Choi YH, Park EM, Kang JL, Kim HS. The phosphodiesterase 10 inhibitor papaverine exerts anti-inflammatory and neuroprotective effects via the PKA signaling pathway in neuroinflammation and Parkinson's disease mouse models. J Neuroinflammation 2019; 16:246. [PMID: 31791357 PMCID: PMC6888927 DOI: 10.1186/s12974-019-1649-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023] Open
Abstract
Background Neuroinflammation plays a pivotal role in the pathogenesis of Parkinson’s disease (PD). Thus, the development of agents that can control neuroinflammation has been suggested as a promising therapeutic strategy for PD. In the present study, we investigated whether the phosphodiesterase (PDE) 10 inhibitor has anti-inflammatory and neuroprotective effects in neuroinflammation and PD mouse models. Methods Papaverine (PAP) was utilized as a selective inhibitor of PDE10. The effects of PAP on the expression of pro-inflammatory molecules were examined in lipopolysaccharide (LPS)–stimulated BV2 microglial cells by ELISA, RT-PCR, and Western blot analysis. The effects of PAP on transcription factors were analyzed by the electrophoretic mobility shift assay, the reporter gene assay, and Western blot analysis. Microglial activation and the expression of proinflammatory molecules were measured in the LPS- or MPTP-injected mouse brains by immunohistochemistry and RT-PCR analysis. The effect of PAP on dopaminergic neuronal cell death and neurotrophic factors were determined by immunohistochemistry and Western blot analysis. To assess mouse locomotor activity, rotarod and pole tests were performed in MPTP-injected mice. Results PAP inhibited the production of nitric oxide and proinflammatory cytokines in LPS-stimulated microglia by modulating various inflammatory signals. In addition, PAP elevated intracellular cAMP levels and CREB phosphorylation. Treatment with H89, a PKA inhibitor, reversed the anti-inflammatory effects of PAP, suggesting the critical role of PKA signaling in the anti-inflammatory effects of PAP. We verified the anti-inflammatory effects of PAP in the brains of mice with LPS-induced systemic inflammation. PAP suppressed microglial activation and proinflammatory gene expression in the brains of these mice, and these effects were reversed by H89 treatment. We further examined the effects of PAP on MPTP-injected PD model mice. MPTP-induced dopaminergic neuronal cell death and impaired locomotor activity were recovered by PAP. In addition, PAP suppressed microglial activation and proinflammatory mediators in the brains of MPTP-injected mice. Conclusions PAP has strong anti-inflammatory and neuroprotective effects and thus may be a potential candidate for treating neuroinflammatory disorders such as PD.
Collapse
Affiliation(s)
- Yu-Young Lee
- Department of Molecular Medicine and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Jin-Sun Park
- Department of Molecular Medicine and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Yea-Hyun Leem
- Department of Molecular Medicine and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Jung-Eun Park
- Department of Molecular Medicine and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-dong, Gangseo-gu, Seoul, 07804, South Korea
| | - Youn-Hee Choi
- Department of Physiology and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Eun-Mi Park
- Department of Pharmacology and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea.,Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Jihee Lee Kang
- Department of Physiology and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 808-1 Magok-dong, Gangseo-gu, Seoul, 07804, South Korea. .,Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
26
|
Activation of TGR5 Partially Alleviates High Glucose-Induced Cardiomyocyte Injury by Inhibition of Inflammatory Responses and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6372786. [PMID: 31871553 PMCID: PMC6906824 DOI: 10.1155/2019/6372786] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/23/2019] [Indexed: 12/17/2022]
Abstract
High glucose- (HG-) induced cardiomyocyte injury is the leading cause of diabetic cardiomyopathy, which is associated with the induction of inflammatory responses and oxidative stress. TGR5 plays an important role in the regulation of glucose metabolism. However, whether TGR5 has cardioprotective effects against HG-induced cardiomyocyte injury is unknown. Neonatal mouse cardiomyocytes were isolated and incubated in a HG medium. Protein and mRNA expression was detected by western blotting and RT-PCR, respectively. Cell apoptosis was determined by Hoechst 33342 staining and flow cytometry. After treatment of cells with HG, TGR5-selective agonist INT-777 reduced the increase in expression of proinflammatory cytokines and NF-κB, whereas pretreatment of cells with TGR5 shRNA significantly reduced the inhibitory effects of INT-777. We also found that INT-777 increased the protein expression of Nrf2 and HO-1. In the presence of TGR5 shRNA, the expression of Nrf2 and HO-1 was reduced, indicating that TGR5 may exert an antioxidant effect partially through the Nrf2/HO-1 pathway. Furthermore, INT-777 treatment inhibited HG-induced ROS production and apoptosis that were attenuated in the presence of TGR5 shRNA or ZnPP (HO-1 inhibitor). Activation of TGR5 has cardioprotective effects against HG-induced cardiomyocyte injury and could be a pharmacological target for the treatment of diabetic cardiomyopathy.
Collapse
|
27
|
Pulsed Electromagnetic Fields Reduce Interleukin-6 Expression in Intervertebral Disc Cells Via Nuclear Factor-κβ and Mitogen-Activated Protein Kinase p38 Pathways. Spine (Phila Pa 1976) 2019; 44:E1290-E1297. [PMID: 31689248 DOI: 10.1097/brs.0000000000003136] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN This is an in vitro study of bovine disc cells exposed to pulsed electromagnetic fields. OBJECTIVE The purpose of the present study was to investigate whether pulsed electromagnetic fields (PEMF) effects on the expression of interleukin-6 (IL-6) expression is mediated by two known inflammation regulators, nuclear factor-κB (NF-κβ) and phosphorylated mitogen-activated protein kinase p38 (p38-MAPK) signaling pathways SUMMARY OF BACKGROUND DATA.: Inflammatory cytokines play a dominant role in the pathogenesis of disc degeneration. Increasing evidence showed that PEMF, a noninvasive biophysical stimulation, can have physiologically beneficial effects on inflammation and tissue repair. Our previous research shows that PEMF treatment can reduce IL-6 expression by intervertebral disc cells. However, the underlying mechanisms of PEMF action are yet to be uncovered. METHODS Intervertebral disc nuclear pulposus cells were challenged with interleukin-1α (IL-1α) (for mimicking inflammatory microenvironment) and treated with PEMF simultaneously up to 4 hours. Cells were then collected for NF-κβ and phosphorylated p38-MAPK protein detection with Western blot. Additionally, the RelA (p65) subunit of NF-κβ was examined with immunostaining for assessment of NF-κβ activation. RESULTS As expected, Western blot results showed that both NF-κβ and phosphorylated p38 expression were significantly increased by IL-1α treatment. This induction was significantly inhibited to control condition levels by PEMF treatment. Immunostaining demonstrated similar trends, that PEMF treatment reduced the NF-κβ activation induced by IL-1α exposure. CONCLUSION Our data indicate that the previously-reported inhibitory effect of PEMF treatment on disc inflammation is mediated by NF-κβ and phosphorylated p38-MAPK signaling pathways. These results further establish PEMFs anti-inflammatory activity, and may inform potential future clinical uses for management of inflammation associated with disc degeneration. LEVEL OF EVIDENCE N/A.
Collapse
|
28
|
Wang Z, Liang Y, Zhang L, Zhang N, Liu Q, Wang Z. Phosphodiesterase 4 inhibitor activates AMPK-SIRT6 pathway to prevent aging-related adipose deposition induced by metabolic disorder. Aging (Albany NY) 2019; 10:2394-2406. [PMID: 30227388 PMCID: PMC6188481 DOI: 10.18632/aging.101559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/12/2018] [Indexed: 12/16/2022]
Abstract
Rolipram is a selective phosphodiesterase 4 (PDE4) inhibitor that exerts a variety of effects, including anti-inflammatory, immunosuppressive, and anti-tumor effects. The aim of this study was to investigate the effect of rolipram on metabolic disorder and its underlying mechanisms. Metabolic disorder was induced in 8-week-old wild type BABL/c mice by administration of D-galactose for 4 weeks. Simultaneously the mice were administered vehicle or rolipram. Alternatively, beginning at 3 or 21 months, the mice were administered db-cAMP for 3 months, with or without a high-fat-diet (HFD) to induce metabolic disorder. In both models, better metabolic function was observed in rolipram-treated mice. Rolipram reduced adipose deposition and inflammation and reserved metabolic disorder. Treatment with rolipram increased the AMPK phosphorylation and SIRT6 levels in the liver and kidney while reducing NF-κB acetylation. In vitro, these effects were blocked by suppression of SIRT6 expression using specific siRNA. Increased cAMP levels reduced excessive adipose deposition, and improved adipose distribution in presenile mice. These findings provide a promising strategy for the treatment of aging-related metabolic dysfunctions and suggest that selective PDE4 inhibitors may be useful agents for the treatment of aging-related metabolic diseases.
Collapse
Affiliation(s)
- Zhuoran Wang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yaru Liang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Lu Zhang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Nannan Zhang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Qingfei Liu
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Zhao Wang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
29
|
Dubey NK, Wei HJ, Yu SH, Williams DF, Wang JR, Deng YH, Tsai FC, Wang PD, Deng WP. Adipose-derived Stem Cells Attenuates Diabetic Osteoarthritis via Inhibition of Glycation-mediated Inflammatory Cascade. Aging Dis 2019; 10:483-496. [PMID: 31164994 PMCID: PMC6538220 DOI: 10.14336/ad.2018.0616] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/16/2018] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is well-known to exert complications such as retinopathy, cardiomyopathy and neuropathy. However, in recent years, an elevated osteoarthritis (OA) complaints among diabetics have been observed, portending the risk of diabetic OA. Since formation of advanced glycation end products (AGE) is believed to be the etiology of various diseases under hyperglycemic conditions, we firstly established that streptozotocin-induced DM could potentiate the development of OA in C57BL/6J mouse model, and further explored the intra-articularly administered adipose-derived stem cell (ADSC) therapy focusing on underlying AGE-associated mechanism. Our results demonstrated that hyperglycemic mice exhibited OA-like structural impairments including a proteoglycan loss and articular cartilage fibrillations in knee joint. Highly expressed levels of carboxymethyl lysine (CML), an AGE and their receptors (RAGE), which are hallmarks of hyperglycemic microenvironment were manifested. The elevated oxidative stress in diabetic OA knee-joint was revealed through increased levels of malondialdehyde (MDA). Further, oxidative stress-activated nuclear factor kappa B (NF-κB), the marker of proinflammatory signalling pathway was also accrued; and levels of matrix metalloproteinase-1 and 13 were upregulated. However, ADSC treatment attenuated all OA-like changes by 4 weeks, and dampened levels of CML, RAGE, MDA, NF-κB, MMP-1 and 13. These results suggest that during repair and regeneration, ADSCs inhibited glycation-mediated inflammatory cascade and rejuvenated cartilaginous tissue, thereby promoting knee-joint integrity in diabetic milieu.
Collapse
Affiliation(s)
- Navneet Kumar Dubey
- 1Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,2Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Jian Wei
- 2Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,3School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,4School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sung-Hsun Yu
- 2Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - David F Williams
- 5Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, USA
| | - Joseph R Wang
- 6Department of Periodontics, College of Dental Medicine, Columbia University, New York, USA
| | - Yue-Hua Deng
- 7Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Feng-Chou Tsai
- 8Stem Cell Research Center, Cosmetic Clinic Group, Taipei, Taiwan
| | - Peter D Wang
- 4School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,9Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Win-Ping Deng
- 2Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,4School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,10Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
30
|
Chen S, Zhou H, Zhang G, Meng J, Deng K, Zhou W, Wang H, Wang Z, Hu N, Suo Y. Anthocyanins from Lycium ruthenicum Murr. Ameliorated d-Galactose-Induced Memory Impairment, Oxidative Stress, and Neuroinflammation in Adult Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3140-3149. [PMID: 30813721 DOI: 10.1021/acs.jafc.8b06402] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Lycium ruthenicum Murr. (LR) is a perennial shrub commonly used as a nutritional food and medicine. Herein, we identified 12 anthocyanins from LR, with petunidin derivatives constituting approximately 97% of the total anthocyanin content. Furthermore, the potential mechanism of anthocyanins exerting neuroprotective effects in d-galactose (d-gal)-treated rats was explored. Behavioral results showed that anthocyanins relieved d-gal-induced memory disorder. Additionally, anthocyanins reduced receptor for advanced glycation end products (RAGE) and suppressed oxidative stress caused by d-gal. Anthocyanins suppressed microgliosis and astrocytosis and reduced the overexpression of nuclear factor kappa B (NF-κB), interleukin-1-β (IL-1β), cyclooxygenase-2 (COX-2), and tumor necrosis factor-α (TNF-α). Moreover, anthocyanins lowered C-jun N-terminal kinase ( p-JNK), caspase-3 levels, and the B-cell lymphoma 2-associated X protein/B-cell lymphoma 2 (Bax/Bcl-2) ratio. Thus, anthocyanins from LR attenuated memory disfunction, neuroinflammation, and neurodegeneration caused by d-gal, possibly through the RAGE/NF-κB/JNK pathway, representing a promising, safe candidate for prevention and therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shasha Chen
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Haonan Zhou
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Gong Zhang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Jing Meng
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Kai Deng
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Wu Zhou
- State Key Laboratory of Plateau Ecology and Agriculture , Qinghai University , Xining 810016 , China
| | - Honglun Wang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- State Key Laboratory of Plateau Ecology and Agriculture , Qinghai University , Xining 810016 , China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research , Xining , 810001 , People's Republic of China
| | - Zhenhua Wang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
| | - Na Hu
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- State Key Laboratory of Plateau Ecology and Agriculture , Qinghai University , Xining 810016 , China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research , Xining , 810001 , People's Republic of China
| | - Yourui Suo
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology , Chinese Academy of Sciences , Xining , 810008 , China
- State Key Laboratory of Plateau Ecology and Agriculture , Qinghai University , Xining 810016 , China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research , Xining , 810001 , People's Republic of China
| |
Collapse
|
31
|
Borst K, Schwabenland M, Prinz M. Microglia metabolism in health and disease. Neurochem Int 2018; 130:104331. [PMID: 30423423 DOI: 10.1016/j.neuint.2018.11.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/24/2018] [Accepted: 11/08/2018] [Indexed: 12/11/2022]
Abstract
In the last decade tremendous progress has been made in understanding how the immune system reacts to insults. During this progress it became obvious that those immune responses are tightly regulated and cross-linked with distinct metabolic changes in immune cells. Extensive research has been conducted mainly on subtypes of T cells, which use different metabolic pathways during differentiation processes and activation states. In addition, it has also been established later, that the innate immune cell lineage of myeloid cells includes a variety of different subsets of bone marrow-derived as well as tissue-specific macrophages, which elicit much more functions than simply killing bacteria. To execute this high variety of functions, also macrophages use different metabolic pathways and are tightly regulated by key metabolic regulators, such as the mechanistic target of rapamycin (mTOR). Upon activation, metabolic changes within the cell occur to meet the requirements of the phenotypic switch. In addition, metabolic changes correlate with the ability of innate immune cells to show hallmarks of adaptive immune responses. Little is known about specific metabolic changes of myeloid cells and specifically microglia in vivo. Microglia are key players in neurodegenerative and neuroinflammatory diseases and have become a major target of medical research. Here, we review the existing data on microglia metabolism and the connection of microglia phenotypes with neuroinflammatory and neurodegenerative diseases. Lastly, we will discuss how our knowledge about the cellular metabolism might be used to develop new treatment options for neurological diseases.
Collapse
Affiliation(s)
- Katharina Borst
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Germany
| | - Marius Schwabenland
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Germany.
| |
Collapse
|
32
|
Wiejak J, van Basten B, Luchowska-Stańska U, Hamilton G, Yarwood SJ. The novel exchange protein activated by cyclic AMP 1 (EPAC1) agonist, I942, regulates inflammatory gene expression in human umbilical vascular endothelial cells (HUVECs). BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:264-276. [PMID: 30414891 PMCID: PMC6325792 DOI: 10.1016/j.bbamcr.2018.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022]
Abstract
Exchange protein activated by cyclic AMP (EPAC1) suppresses multiple inflammatory actions in vascular endothelial cells (VECs), partly due to its ability to induce expression of the suppressor of cytokine signalling 3 (SOCS3) gene, the protein product of which inhibits interleukin 6 (IL6) signalling through the JAK/STAT3 pathway. Here, for the first time, we use the non-cyclic nucleotide EPAC1 agonist, I942, to determine its actions on cellular EPAC1 activity and cyclic AMP-regulated gene expression in VECs. We demonstrate that I942 promotes EPAC1 and Rap1 activation in HEK293T cells and induces SOCS3 expression and suppresses IL6-stimulated JAK/STAT3 signalling in HUVECs. SOCS3 induction by I942 in HUVECs was blocked by the EPAC1 antagonist, ESI-09, and EPAC1 siRNA, but not by the broad-spectrum protein kinase A (PKA) inhibitor, H89, indicating that I942 regulates SOCS3 gene expression through EPAC1. RNA sequencing was carried out to further identify I942-regulated genes in HUVECs. This identified 425 I942-regulated genes that were also regulated by the EPAC1-selective cyclic AMP analogue, 007, and the cyclic AMP-elevating agents, forskolin and rolipram (F/R). The majority of genes identified were suppressed by I942, 007 and F/R treatment and many were involved in the control of key vascular functions, including the gene for the cell adhesion molecule, VCAM1. I942 and 007 also inhibited IL6-induced expression of VCAM1 at the protein level and blocked VCAM1-dependent monocyte adhesion to HUVECs. Overall, I942 represents the first non-cyclic nucleotide EPAC1 agonist in cells with the ability to suppress IL6 signalling and inflammatory gene expression in VECs. The novel EPAC1 ligand I942 activates cellular EPAC1 and Rap1 GTPase. I942 induces SOCS3 gene expression in vascular endothelial cells (VECs). I942 suppresses JAK/STAT3 signalling from the IL6 receptor in VECs. I942 regulates 425 novel gene targets in VECs. I942 suppresses VCAM1 expression and monocyte adhesion in VECs.
Collapse
Affiliation(s)
- Jolanta Wiejak
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Boy van Basten
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Urszula Luchowska-Stańska
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Graham Hamilton
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, University of Glasgow, Bearsden G61 1QH, UK
| | - Stephen J Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh Campus, Edinburgh EH14 4AS, UK.
| |
Collapse
|
33
|
Cai B, Seong KJ, Bae SW, Chun C, Kim WJ, Jung JY. A synthetic diosgenin primary amine derivative attenuates LPS-stimulated inflammation via inhibition of NF-κB and JNK MAPK signaling in microglial BV2 cells. Int Immunopharmacol 2018; 61:204-214. [PMID: 29890414 DOI: 10.1016/j.intimp.2018.05.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 02/07/2023]
Abstract
Diosgenin, a precursor of steroid hormones in plants, is known to exhibit diverse pharmacological activities including anti-inflammatory properties. In this study, (3β, 25R)‑spirost‑5‑en‑3‑oxyl (2‑((2((2‑aminoethyl)amino)ethyl)amino)ethyl) carbamate (DGP), a new synthetic diosgenin derivative incorporating primary amine was used to investigate its anti-inflammatory effects and underlying mechanisms of action in lipopolysaccharide (LPS)-stimulated microglial BV2 cells. Pretreatment with DGP resulted in significant inhibition of nitric oxide (NO) synthesis, and down-regulation of nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-stimulated microglial BV2 cells. In addition, DGP decreased the production of reactive oxygen species (ROS) and pro-inflammatory cytokines such as interleukin (IL)-6, IL-1β, and tumor necrosis factor alpha (TNF-α). The inhibitory effects of DGP on these inflammatory mediators in LPS-stimulated microglial BV2 cells were regulated by NF-κB signaling through blocking p65 nuclear translocation and NF-κB p65/DNA binding activity. DGP also blocked the phosphorylation of c-Jun amino-terminal kinase (JNK), but not p38 kinase or extracellular signal-regulated kinases (ERK). The NF-κB inhibitor JSH-23 and JNK-specific inhibitor SP600125 significantly decreased NO production and IL-6 release in LPS-stimulated BV2 cells, respectively. The overall results demonstrate that DGP has anti-inflammatory effects on LPS-stimulated BV2 cells via inhibition of NF-κB and JNK activation, suggesting that DGP is a potential prophylactic agent in various neurodegenerative disorders.
Collapse
Affiliation(s)
- Bangrong Cai
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Kyung-Joo Seong
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Sun-Woong Bae
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Changju Chun
- Research Institute of Drug Development, College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Won-Jae Kim
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| | - Ji-Yeon Jung
- Dental Science Research Institute, Department of Oral Physiology, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
34
|
Pan Y, Xu C, Pan ZK. MKP-1 negative regulates Staphylococcus aureus induced inflammatory responses in Raw264.7 cells: roles of PKA-MKP-1 pathway and enhanced by rolipram. Sci Rep 2017; 7:12366. [PMID: 28959039 PMCID: PMC5620070 DOI: 10.1038/s41598-017-10187-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/02/2017] [Indexed: 11/22/2022] Open
Abstract
MAP phosphatases (MKP)-1 acts as an important regulator of innate immune response through a mechanism of control and attention both MAPK and NF-κB molecules during bacterial infection. However, the regulatory role of MKP-1 in the interplay between MAPK and NFκB pathway molecules is still not fully understood. In present study, we showed a direct interactions of p38, ERK or IκBα with MKP-1, and demonstrated that MKP-1 was a pivotal feedback control for both MAP kinases and NF-κB pathway in response to S. aureus. In addition, we found that rolipram had anti-inflammatory activity and repressed IκBα activation induced by S. aureus via PKA-MKP-1 pathway. Our report also demonstrated that PKA-cα can directly bind to IκBα upon S. aureus stimulation, which influenced the downstream signaling of PKA pathway, including altered the expression of MKP-1. These results presented a novel mechanism of PKA and IκB pathway, which may be targeted for treating S. aureus infection.
Collapse
Affiliation(s)
- Yiqing Pan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
- Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai, 200025, China
| | - Chen Xu
- Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai, 200025, China.
| | - Zhixing K Pan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, Toledo, OH, 43614, USA.
| |
Collapse
|
35
|
Inhibition of PDE2 reverses beta amyloid induced memory impairment through regulation of PKA/PKG-dependent neuro-inflammatory and apoptotic pathways. Sci Rep 2017; 7:12044. [PMID: 28935920 PMCID: PMC5608906 DOI: 10.1038/s41598-017-08070-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/29/2017] [Indexed: 11/08/2022] Open
Abstract
Beta amyloid peptides (Aβ) are known risk factors involved in cognitive impairment, neuroinflammatory and apoptotic processes in Alzheimer’s disease (AD). Phosphodiesterase 2 (PDE2) inhibitors increase the intracellular cAMP and/or cGMP activities, which may ameliorate cognitive deficits associated with AD. However, it remains unclear whether PDE2 mediated neuroapoptotic and neuroinflammatory events, as well as cognitive performance in AD are related to cAMP/cGMP-dependent pathways. The present study investigated how the selective PDE2 inhibitor BAY60-7550 (BAY) affected Aβ-induced learning and memory impairment in two classic rodent models. IL-22 and IL-17, Bax and Bcl-2, PKA/PKG and the brain derived neurotropic factor (BDNF) levels in hippocampus and cortex were detected with immunoblotting assay. The results showed that BAY reversed Aβ-induced cognitive impairment as shown in the water maze test and step-down test. Moreover, BAY treatment reversed the Aβ-induced changes in IL-22 and IL-17 and the ratio of Bax/Bcl-2. Changes in cAMP/cGMP levels, PKA/PKG and BDNF expression were also prevented by BAY. These effects of BAY on memory performance and related neurochemical changes were partially blocked by the PKG inhibitor KT 5823. These findings indicated that the protective effects of BAY against Aβ-induced memory deficits might involve the regulation of neuroinflammation and neuronal apoptotic events.
Collapse
|
36
|
Avila DV, Myers SA, Zhang J, Kharebava G, McClain CJ, Kim HY, Whittemore SR, Gobejishvili L, Barve S. Phosphodiesterase 4b expression plays a major role in alcohol-induced neuro-inflammation. Neuropharmacology 2017; 125:376-385. [PMID: 28807677 DOI: 10.1016/j.neuropharm.2017.08.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/09/2017] [Indexed: 02/07/2023]
Abstract
It is increasingly evident that alcohol-induced, gut-mediated peripheral endotoxemia plays a significant role in glial cell activation and neuro-inflammation. Using a mouse model of chronic alcohol feeding, we examined the causal role of endotoxin- and cytokine-responsive Pde4 subfamily b (Pde4b) expression in alcohol-induced neuro-inflammation. Both pharmacologic and genetic approaches were used to determine the regulatory role of Pde4b. In C57Bl/6 wild type (WT) alcohol fed (WT-AF) animals, alcohol significantly induced peripheral endotoxemia and Pde4b expression in brain tissue, accompanied by a decrease in cAMP levels. Further, along with Pde4b, there was a robust activation of astrocytes and microglia accompanied by significant increases in the inflammatory cytokines (Tnfα, Il-1β, Mcp-1 and Il-17) and the generalized inflammatory marker Cox-2. At the cellular level, alcohol and inflammatory mediators, particularly LPS, Tnfα and Hmgb1 significantly activated microglial cells (Iba-1 expression) and selectively induced Pde4b expression with a minimal to no change in Pde4a and d isoforms. In comparison, the alcohol-induced decrease in brain cAMP levels was completely inhibited in WT mice treated with the Pde4 specific pharmacologic inhibitor rolipram and in Pde4b-/- mice. Moreover, all the observed markers of alcohol-induced brain inflammation were markedly attenuated. Importantly, glial cell activation induced by systemic endotoxemia (LPS administration) was also markedly decreased in Pde4b-/- mice. Taken together, these findings strongly support the notion that Pde4b plays a critical role in coordinating alcohol-induced, peripheral endotoxemia mediated neuro-inflammation and could serve as a significant therapeutic target.
Collapse
Affiliation(s)
- Diana V Avila
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA; Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Scott A Myers
- Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, USA; Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - JingWen Zhang
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA; Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Giorgi Kharebava
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, MD, USA
| | - Craig J McClain
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA; Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, MD, USA
| | - Scott R Whittemore
- Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, USA; Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA; Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Leila Gobejishvili
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA; Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Shirish Barve
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA; Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
37
|
Gnanasegaran N, Govindasamy V, Mani V, Abu Kasim NH. Neuroimmunomodulatory properties of DPSCs in anin vitromodel of Parkinson's disease. IUBMB Life 2017; 69:689-699. [DOI: 10.1002/iub.1655] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/16/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Nareshwaran Gnanasegaran
- Department of Restorative Dentistry; Faculty of Dentistry, University of Malaya; Kuala Lumpur Malaysia
| | - Vijayendran Govindasamy
- Department of Restorative Dentistry; Faculty of Dentistry, University of Malaya; Kuala Lumpur Malaysia
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology; College of Pharmacy, Qassim University; Buraidah Kingdom of Saudi Arabia
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry; Faculty of Dentistry, University of Malaya; Kuala Lumpur Malaysia
| |
Collapse
|
38
|
Song GJ, Suk K. Pharmacological Modulation of Functional Phenotypes of Microglia in Neurodegenerative Diseases. Front Aging Neurosci 2017; 9:139. [PMID: 28555105 PMCID: PMC5430023 DOI: 10.3389/fnagi.2017.00139] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/25/2017] [Indexed: 01/06/2023] Open
Abstract
Microglia are the resident innate immune cells of the central nervous system that mediate brain homeostasis maintenance. Microglia-mediated neuroinflammation is a hallmark shared by various neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis. Numerous studies have shown microglial activation phenotypes to be heterogeneous; however, these microglial phenotypes can largely be categorized as being either M1 or M2 type. Although the specific classification of M1 and M2 functionally polarized microglia remains a topic for debate, the use of functional modulators of microglial phenotypes as potential therapeutic approaches for the treatment of neurodegenerative diseases has garnered considerable attention. This review discusses M1 and M2 microglial phenotypes and their relevance in neurodegenerative disease models, as described in recent literature. The modulation of microglial polarization toward the M2 phenotype may lead to development of future therapeutic and preventive strategies for neuroinflammatory and neurodegenerative diseases. Thus, we focus on recent studies of microglial polarization modulators, with a particular emphasis on the small-molecule compounds and their intracellular target proteins.
Collapse
Affiliation(s)
- Gyun Jee Song
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, Brain Science & Engineering Institute, School of Medicine, Kyungpook National UniversityDaegu, South Korea
| | - Kyoungho Suk
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, Brain Science & Engineering Institute, School of Medicine, Kyungpook National UniversityDaegu, South Korea
| |
Collapse
|
39
|
Sanguinarine protects against osteoarthritis by suppressing the expression of catabolic proteases. Oncotarget 2017; 8:62900-62913. [PMID: 28968958 PMCID: PMC5609890 DOI: 10.18632/oncotarget.17036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/24/2017] [Indexed: 12/17/2022] Open
Abstract
Inflammatory cytokines play critical roles in the pathogenesis of osteoarthritis. Recent studies have demonstrated that natural active substances can serve as alternative therapeutic agents for the prevention and treatment of osteoarthritis. Sanguinarine, an alkaloid isolated from the roots of Sanguinaria canadensis, is known to have anti-inflammatory properties. The aim of the present study was to investigate the therapeutic effect of Sanguinarine against osteoarthritis. Sanguinarine inhibited interleukin-1β-induced expression of matrix metalloproteinase 1, 3, and 13, and A disintegrin and metalloproteinase with thrombospondin motifs-5 in chondrocytes, which involved the nuclear factor-κB and c-Jun N-terminal kinase signalling pathways. Furthermore, the study of interleukin-1β-induced cartilage matrix degradation in an anterior cruciate ligament transection-induced osteoarthritis model revealed that Sanguinarine ameliorated osteoarthritis by inhibiting the expression of matrix metalloproteinase 1, 3, and 13, and A disintegrin and metalloproteinase with thrombospondin motifs-5. In conclusion, we demonstrated for the first time that Sanguinarine suppressed the expression of matrix metalloproteinase 1, 3, and 13, and A disintegrin and metalloproteinase with thrombospondin motifs-5 in vitro, ex vivo, and in vivo, indicating its potential usefulness in treating osteoarthritis.
Collapse
|
40
|
Knott EP, Assi M, Rao SNR, Ghosh M, Pearse DD. Phosphodiesterase Inhibitors as a Therapeutic Approach to Neuroprotection and Repair. Int J Mol Sci 2017; 18:E696. [PMID: 28338622 PMCID: PMC5412282 DOI: 10.3390/ijms18040696] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 12/21/2022] Open
Abstract
A wide diversity of perturbations of the central nervous system (CNS) result in structural damage to the neuroarchitecture and cellular defects, which in turn are accompanied by neurological dysfunction and abortive endogenous neurorepair. Altering intracellular signaling pathways involved in inflammation and immune regulation, neural cell death, axon plasticity and remyelination has shown therapeutic benefit in experimental models of neurological disease and trauma. The second messengers, cyclic adenosine monophosphate (cyclic AMP) and cyclic guanosine monophosphate (cyclic GMP), are two such intracellular signaling targets, the elevation of which has produced beneficial cellular effects within a range of CNS pathologies. The only known negative regulators of cyclic nucleotides are a family of enzymes called phosphodiesterases (PDEs) that hydrolyze cyclic nucleotides into adenosine monophosphate (AMP) or guanylate monophosphate (GMP). Herein, we discuss the structure and physiological function as well as the roles PDEs play in pathological processes of the diseased or injured CNS. Further we review the approaches that have been employed therapeutically in experimental paradigms to block PDE expression or activity and in turn elevate cyclic nucleotide levels to mediate neuroprotection or neurorepair as well as discuss both the translational pathway and current limitations in moving new PDE-targeted therapies to the clinic.
Collapse
Affiliation(s)
- Eric P Knott
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Mazen Assi
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Sudheendra N R Rao
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Neuroscience Program, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- Bruce Wayne Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
41
|
Cross-talk between LRRK2 and PKA: implication for Parkinson's disease? Biochem Soc Trans 2017; 45:261-267. [DOI: 10.1042/bst20160396] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 01/16/2023]
Abstract
Evidence indicates that leucine-rich repeat kinase 2 (LRRK2) controls multiple processes in neurons and glia cells. Deregulated LRRK2 activity due to gene mutation represents the most common cause of autosomal dominant Parkinson's disease (PD). Protein kinase A (PKA)-mediated signaling is a key regulator of brain function. PKA-dependent pathways play an important role in brain homeostasis, neuronal development, synaptic plasticity, control of microglia activation and inflammation. On the other hand, a decline of PKA signaling was shown to contribute to the progression of several neurodegenerative diseases, including PD. In this review, we will discuss the accumulating evidence linking PKA and LRRK2 in neuron and microglia functions, and offer an overview of the enigmatic cross-talk between these two kinases with molecular and cellular implications.
Collapse
|
42
|
Maggio DM, Singh A, Iorgulescu JB, Bleicher DH, Ghosh M, Lopez MM, Tuesta LM, Flora G, Dietrich WD, Pearse DD. Identifying the Long-Term Role of Inducible Nitric Oxide Synthase after Contusive Spinal Cord Injury Using a Transgenic Mouse Model. Int J Mol Sci 2017; 18:ijms18020245. [PMID: 28125047 PMCID: PMC5343782 DOI: 10.3390/ijms18020245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/05/2017] [Accepted: 01/15/2017] [Indexed: 02/07/2023] Open
Abstract
Inducible nitric oxide synthase (iNOS) is a potent mediator of oxidative stress during neuroinflammation triggered by neurotrauma or neurodegeneration. We previously demonstrated that acute iNOS inhibition attenuated iNOS levels and promoted neuroprotection and functional recovery after spinal cord injury (SCI). The present study investigated the effects of chronic iNOS ablation after SCI using inos-null mice. iNOS-/- knockout and wild-type (WT) control mice underwent a moderate thoracic (T8) contusive SCI. Locomotor function was assessed weekly, using the Basso Mouse Scale (BMS), and at the endpoint (six weeks), by footprint analysis. At the endpoint, the volume of preserved white and gray matter, as well as the number of dorsal column axons and perilesional blood vessels rostral to the injury, were quantified. At weeks two and three after SCI, iNOS-/- mice exhibited a significant locomotor improvement compared to WT controls, although a sustained improvement was not observed during later weeks. At the endpoint, iNOS-/- mice showed significantly less preserved white and gray matter, as well as fewer dorsal column axons and perilesional blood vessels, compared to WT controls. While short-term antagonism of iNOS provides histological and functional benefits, its long-term ablation after SCI may be deleterious, blocking protective or reparative processes important for angiogenesis and tissue preservation.
Collapse
Affiliation(s)
- Dominic M Maggio
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institute of Heath, Bethesda, MD 20824, USA.
| | - Amanpreet Singh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - J Bryan Iorgulescu
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Drew H Bleicher
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Michael M Lopez
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Luis M Tuesta
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | - Govinder Flora
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
43
|
Park T, Chen H, Kevala K, Lee JW, Kim HY. N-Docosahexaenoylethanolamine ameliorates LPS-induced neuroinflammation via cAMP/PKA-dependent signaling. J Neuroinflammation 2016; 13:284. [PMID: 27809877 PMCID: PMC5096293 DOI: 10.1186/s12974-016-0751-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/24/2016] [Indexed: 01/22/2023] Open
Abstract
Background Brain inflammation has been implicated as a critical mechanism responsible for the progression of neurodegeneration and characterized by glial cell activation accompanied by production of inflammation-related cytokines and chemokines. Growing evidence also suggests that metabolites derived from docosahexaenoic acid (DHA) have anti-inflammatory and pro-resolving effects; however, the possible role of N-docosahexaenoylethanolamine (synaptamide), an endogenous neurogenic and synaptogenic metabolite of DHA, in inflammation, is largely unknown. (The term “synaptamide” instead of “DHEA” was used for N-docosahexaenoylethanolamine since DHEA is a widely used and accepted term for the steroid, dehydroepiandrosterone.) In the present study, we tested this possibility using a lipopolysaccharide (LPS)-induced neuroinflammation model both in vitro and in vivo. Methods For in vitro studies, we used P3 primary rat microglia and immortalized murine microglia cells (BV2) to assess synaptamide effects on LPS-induced cytokine/chemokine/iNOS (inducible nitric oxide synthase) expression by quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). To evaluate in vivo effects, mice were intraperitoneally (i.p.) injected with LPS followed by synaptamide, and expression of proinflammatory mediators was measured by qPCR and western blot analysis. Activation of microglia and astrocyte in the brain was examined by Iba-1 and GFAP immunostaining. Results Synaptamide significantly reduced LPS-induced production of TNF-α and NO in cultured microglia cells. Synaptamide increased intracellular cAMP levels, phosphorylation of PKA, and phosphorylation of CREB but suppressed LPS-induced nuclear translocation of NF-κB p65. Conversely, adenylyl cyclase or PKA inhibitors abolished the synaptamide effect on p65 translocation as well as TNF-α and iNOS expression. Administration of synaptamide following LPS injection (i.p.) significantly reduced neuroinflammatory responses, such as microglia activation and mRNA expression of inflammatory cytokines, chemokine, and iNOS in the brain. Conclusions DHA-derived synaptamide is a potent suppressor of neuroinflammation in an LPS-induced model, by enhancing cAMP/PKA signaling and inhibiting NF-κB activation. The anti-inflammatory capability of synaptamide may provide a new therapeutic avenue to ameliorate the inflammation-associated neurodegenerative conditions. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0751-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taeyeop Park
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Huazhen Chen
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Karl Kevala
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Ji-Won Lee
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA. .,National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rm. 3N-07, Bethesda, MD, 20892-9410, USA.
| |
Collapse
|
44
|
Yulia A, Singh N, Lei K, Sooranna SR, Johnson MR. Cyclic AMP Effectors Regulate Myometrial Oxytocin Receptor Expression. Endocrinology 2016; 157:4411-4422. [PMID: 27673556 DOI: 10.1210/en.2016-1514] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The factors that initiate human labor are poorly understood. We have tested the hypothesis that a decline in cAMP/protein kinase A (PKA) function leads to the onset of labor. Initially, we identified myometrial cAMP/PKA-responsive genes (six up-regulated and five down-regulated genes) and assessed their expression in myometrial samples taken from different stages of pregnancy and labor. We found that the oxytocin receptor (OTR) was one of the cAMP-repressed genes, and, given the importance of OTR in the labor process, we studied the mechanisms involved in greater detail using small interfering RNA, chemical agonists, and antagonists of the cAMP effectors. We found that cAMP-repressed genes, including OTR, increased with the onset of labor. Our in vitro studies showed that cAMP acting via PKA reduced OTR expression but that in the absence of PKA, cAMP acts via exchange protein activated by cAMP (EPAC) to increase OTR expression. In early labor myometrial samples, PKA levels and activity declined and Epac1 levels increased, perhaps accounting for the increase in myometrial OTR mRNA and protein levels at this time. In vitro exposure of myometrial cells to stretch and IL-1β increased OTR levels and reduced basal and forskolin-stimulated cAMP and PKA activity, as judged by phospho-cAMP response element-binding protein levels, but neither stretch nor IL-1β had any effect on PKA or EPAC1 levels. In summary, there is a reduction in the activity of the cAMP/PKA pathway with the onset of human labor potentially playing a critical role in regulating OTR expression and the transition from myometrial quiescence to activation.
Collapse
Affiliation(s)
- Angela Yulia
- Chelsea and Westminster Hospital (A.Y., N.S., K.L., S.R.S., M.R.J.), London SW10 9NH, United Kingdom; and Institute of Reproductive and Developmental Biology (A.Y., N.S., K.L., S.R.S., M.R.J.), London W12 0NN, United Kingdom
| | - Natasha Singh
- Chelsea and Westminster Hospital (A.Y., N.S., K.L., S.R.S., M.R.J.), London SW10 9NH, United Kingdom; and Institute of Reproductive and Developmental Biology (A.Y., N.S., K.L., S.R.S., M.R.J.), London W12 0NN, United Kingdom
| | - Kaiyu Lei
- Chelsea and Westminster Hospital (A.Y., N.S., K.L., S.R.S., M.R.J.), London SW10 9NH, United Kingdom; and Institute of Reproductive and Developmental Biology (A.Y., N.S., K.L., S.R.S., M.R.J.), London W12 0NN, United Kingdom
| | - Suren R Sooranna
- Chelsea and Westminster Hospital (A.Y., N.S., K.L., S.R.S., M.R.J.), London SW10 9NH, United Kingdom; and Institute of Reproductive and Developmental Biology (A.Y., N.S., K.L., S.R.S., M.R.J.), London W12 0NN, United Kingdom
| | - Mark R Johnson
- Chelsea and Westminster Hospital (A.Y., N.S., K.L., S.R.S., M.R.J.), London SW10 9NH, United Kingdom; and Institute of Reproductive and Developmental Biology (A.Y., N.S., K.L., S.R.S., M.R.J.), London W12 0NN, United Kingdom
| |
Collapse
|
45
|
Thymoquinone Inhibits IL-1β-Induced Inflammation in Human Osteoarthritis Chondrocytes by Suppressing NF-κB and MAPKs Signaling Pathway. Inflammation 2016; 38:2235-41. [PMID: 26156811 DOI: 10.1007/s10753-015-0206-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Thymoquinone, an active ingredient isolated from Nigella sativa, has been reported to have anti-inflammatory effects. However, the anti-inflammatory effect of thymoquinone on IL-1β-stimulated osteoarthritis chondrocytes remains unclear. In this study, we designed to investigate the anti-inflammatory effects and elucidated the underlying mechanism of thymoquinone on IL-1β-stimulated human osteoarthritis chondrocytes. The effects of thymoquinone on inflammatory mediators COX-2, iNOS, NO, PGE2, as well as MMP-1, MMP3, MMP13 production were detected. The results demonstrated that thymoquinone concentration-dependently inhibited IL-1β-induced COX-2, iNOS, NO, and PGE2 production. Thymoquinone also suppressed IL-1β-induced MMP-1, MMP3, and MMP13 production. We found that thymoquinone significantly inhibited IL-1β-induced NF-κB activation and IκBα degradation. In addition, thymoquinone was found to suppress IL-1β-induced mitogen-activated protein kinases (MAPKs) activation. In conclusion, thymoquinone inhibited IL-1β-induced inflammatory mediator production by inhibition of NF-κB and MAPKs signaling pathways in osteoarthritis chondrocytes. Thymoquinone may be a potential agent in the treatment of osteoarthritis.
Collapse
|
46
|
Pearse DD, Hughes ZA. PDE4B as a microglia target to reduce neuroinflammation. Glia 2016; 64:1698-709. [PMID: 27038323 DOI: 10.1002/glia.22986] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/08/2016] [Accepted: 03/11/2016] [Indexed: 12/12/2022]
Abstract
The importance of microglia in immune homeostasis within the brain is undisputed. Their role in a diversity of neurological and psychiatric diseases as well as CNS injury is the subject of much investigation. Cyclic adenosine monophosphate (AMP) is a critical regulator of microglia homeostasis; as the predominant negative modulator of cyclic AMP signaling within microglia, phosphodiesterase 4 (PDE4) represents a promising target for modulating immune function. PDE4 expression is regulated by inflammation, and in turn, PDE4 inhibition can alter microglia reactivity. As the prototypic PDE4 inhibitor, rolipram, was tested clinically in the 1980s, drug discovery and clinical development of PDE4 inhibitors have been severely hampered by tolerability issues involving nausea and emesis. The two PDE4 inhibitors approved for peripheral inflammatory disorders (roflumilast and apremilast) lack brain penetration and are dose-limited by side effects making them unsuitable for modulating microglial function. Subtype selective inhibitors targeting PDE4B are of high interest given the critical role PDE4B plays in immune function versus the association of PDE4D with nausea and emesis. The challenges and requirements for successful development of a novel brain-penetrant PDE4B inhibitor are discussed in the context of early clinical development strategies. Furthermore, the challenges of monitoring the state of microglia in vivo are highlighted, including a description of the currently available tools and their limitations. Continued drug discovery efforts to identify safe and well-tolerated, brain-penetrant PDE4 inhibitors are a reflection of the confidence in the rationale for modulation of this target to produce meaningful therapeutic benefit in a wide range of neurological conditions and injury. GLIA 2016;64:1698-1709.
Collapse
Affiliation(s)
- Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida.,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida
| | - Zoë A Hughes
- Neuroscience and Pain Research Unit, Pfizer Global Research, Cambridge, Massachusetts
| |
Collapse
|
47
|
Ghosh M, Xu Y, Pearse DD. Cyclic AMP is a key regulator of M1 to M2a phenotypic conversion of microglia in the presence of Th2 cytokines. J Neuroinflammation 2016; 13:9. [PMID: 26757726 PMCID: PMC4711034 DOI: 10.1186/s12974-015-0463-9] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/17/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Microglia and macrophages play a central role in neuroinflammation. Pro-inflammatory cytokines trigger their conversion to a classically activated (M1) phenotype, sustaining inflammation and producing a cytotoxic environment. Conversely, anti-inflammatory cytokines polarize the cells towards an alternatively activated (M2), tissue reparative phenotype. Elucidation of the signal transduction pathways involved in M1 to M2 phenotypic conversion may provide insight into how the innate immune response can be harnessed during distinct phases of disease or injury to mediate neuroprotection and neurorepair. METHODS Microglial cells (cell line and primary) were subjected to combined cyclic adenosine monophosphate (cyclic AMP) and IL-4, or either alone, in the presence of pro-inflammatory mediators, lipopolysaccharide (LPS), or tumor necrosis factor-α (TNF-α). Their effects on the expression of characteristic markers for M1 and M2 microglia were assessed. Similarly, the M1 and M2 phenotypes of microglia and macrophages within the lesion site were then evaluated following a contusive spinal cord injury (SCI) to the thoracic (T8) spinal cord of rats and mice when the agents were administered systemically. RESULTS It was demonstrated that cyclic AMP functions synergistically with IL-4 to promote M1 to M2 conversion of microglia in culture. The combination of cyclic AMP and IL-4, but neither alone, induced an Arg-1(+)/iNOS(-)cell phenotype with concomitant expression of other M2-specific markers including TG2 and RELM-α. M2-converted microglia showed ameliorated production of pro-inflammatory cytokines (TNF-α and IP-10) and reactive oxygen species, with no alteration in phagocytic properties. M2a conversion required protein kinase A (PKA), but not the exchange protein directly activated by cyclic AMP (EPAC). Systemic delivery of cyclic AMP and IL-4 after experimental SCI also promoted a significant M1 to M2a phenotypic change in microglia and macrophage population dynamics in the lesion. CONCLUSIONS Using primary microglia, microglial cell lines, and experimental models of CNS injury, we demonstrate that cyclic AMP levels are a critical determinant in M1-M2 polarization. High levels of cyclic AMP promoted an Arg-1(+) M2a phenotype when microglia were activated with pro-inflammatory stimuli and Th2 cytokines. Th2 cytokines or cyclic AMP independently did not promote these changes. Phenotypic conversion of microglia provides a powerful new therapeutic approach for altering the balance of cytotoxic to reparative microglia in a diversity of neurological diseases and injury.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. .,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Yong Xu
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. .,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. .,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. .,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|