1
|
Karati D, Mukherjee S, Roy S. A Promising Drug Candidate as Potent Therapeutic Approach for Neuroinflammation and Its In Silico Justification of Chalcone Congeners: a Comprehensive Review. Mol Neurobiol 2024; 61:1873-1891. [PMID: 37801205 DOI: 10.1007/s12035-023-03632-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023]
Abstract
Multiple genetic, environmental, and immunological variables cause neuropsychiatric disorders (NPDs). The induced inflammatory immune response is also connected to the severity and treatment outcomes of various NPDs. These reactions also significantly impact numerous brain functions such as GABAergic signaling and neurotransmitter synthesis through inflammatory cytokines and chemokines. Chalcones (1,3-diaryl-2-propen-1-ones) and their heterocyclic counterparts are flavonoids with various biological characteristics including anti-inflammatory activity. Several pure chalcones have been clinically authorized or studied in humans. Chalcones are favored for their diagnostic and therapeutic efficacy in neuroinflammation due to their tiny molecular size, easy manufacturing, and flexibility for changes to adjust lipophilicity ideal for BBB penetrability. These compounds reached an acceptable plasma concentration and were well-tolerated in clinical testing. As a result, they are attracting increasing attention from scientists. However, chalcones' therapeutic potential remains largely untapped. This paper is aimed at highlighting the causes of neuroinflammation, more potent chalcone congeners, their mechanisms of action, and relevant structure-activity relationships.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, West Bengal, 700091, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
2
|
Xue J, Tao K, Wang W, Wang X. What Can Inflammation Tell Us about Therapeutic Strategies for Parkinson's Disease? Int J Mol Sci 2024; 25:1641. [PMID: 38338925 PMCID: PMC10855787 DOI: 10.3390/ijms25031641] [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: 01/01/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder with a complicated etiology and pathogenesis. α-Synuclein aggregation, dopaminergic (DA) neuron loss, mitochondrial injury, oxidative stress, and inflammation are involved in the process of PD. Neuroinflammation has been recognized as a key element in the initiation and progression of PD. In this review, we summarize the inflammatory response and pathogenic mechanisms of PD. Additionally, we describe the potential anti-inflammatory therapies, including nod-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome inhibition, nuclear factor κB (NF-κB) inhibition, microglia inhibition, astrocyte inhibition, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, the peroxisome proliferator-activated receptor γ (PPARγ) agonist, targeting the mitogen-activated protein kinase (MAPK) pathway, targeting the adenosine monophosphate-activated protein kinase (AMPK)-dependent pathway, targeting α-synuclein, targeting miRNA, acupuncture, and exercise. The review focuses on inflammation and will help in designing new prevention strategies for PD.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| |
Collapse
|
3
|
Berglund R, Cheng Y, Piket E, Adzemovic MZ, Zeitelhofer M, Olsson T, Guerreiro-Cacais AO, Jagodic M. The aging mouse CNS is protected by an autophagy-dependent microglia population promoted by IL-34. Nat Commun 2024; 15:383. [PMID: 38195627 PMCID: PMC10776874 DOI: 10.1038/s41467-023-44556-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
Microglia harness an unutilized health-promoting potential in age-related neurodegenerative and neuroinflammatory diseases, conditions like progressive multiple sclerosis (MS). Our research unveils an microglia population emerging in the cortical brain regions of aging mice, marked by ERK1/2, Akt, and AMPK phosphorylation patterns and a transcriptome indicative of activated autophagy - a process critical for cellular adaptability. By deleting the core autophagy gene Ulk1 in microglia, we reduce this population in the central nervous system of aged mice. Notably, this population is found dependent on IL-34, rather than CSF1, although both are ligands for CSF1R. When aging mice are exposed to autoimmune neuroinflammation, the loss of autophagy-dependent microglia leads to neural and glial cell death and increased mortality. Conversely, microglial expansion mediated by IL-34 exhibits a protective effect. These findings shed light on an autophagy-dependent neuroprotective microglia population as a potential target for treating age-related neuroinflammatory conditions, including progressive MS.
Collapse
Affiliation(s)
- Rasmus Berglund
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| | - Yufei Cheng
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Eliane Piket
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Milena Z Adzemovic
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Manuel Zeitelhofer
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, 171 65, Solna, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Andre Ortlieb Guerreiro-Cacais
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| |
Collapse
|
4
|
Socodato R, Rodrigues-Santos A, Tedim-Moreira J, Almeida TO, Canedo T, Portugal CC, Relvas JB. RhoA balances microglial reactivity and survival during neuroinflammation. Cell Death Dis 2023; 14:690. [PMID: 37863874 PMCID: PMC10589285 DOI: 10.1038/s41419-023-06217-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 09/29/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Microglia are the largest myeloid cell population in the brain. During injury, disease, or inflammation, microglia adopt different functional states primarily involved in restoring brain homeostasis. However, sustained or exacerbated microglia inflammatory reactivity can lead to brain damage. Dynamic cytoskeleton reorganization correlates with alterations of microglial reactivity driven by external cues, and proteins controlling cytoskeletal reorganization, such as the Rho GTPase RhoA, are well positioned to refine or adjust the functional state of the microglia during injury, disease, or inflammation. Here, we use multi-biosensor-based live-cell imaging approaches and tissue-specific conditional gene ablation in mice to understand the role of RhoA in microglial response to inflammation. We found that a decrease in RhoA activity is an absolute requirement for microglial metabolic reprogramming and reactivity to inflammation. However, without RhoA, inflammation disrupts Ca2+ and pH homeostasis, dampening mitochondrial function, worsening microglial necrosis, and triggering microglial apoptosis. Our results suggest that a minimum level of RhoA activity is obligatory to concatenate microglia inflammatory reactivity and survival during neuroinflammation.
Collapse
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
| | - Artur Rodrigues-Santos
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Joana Tedim-Moreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Tiago O Almeida
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Teresa Canedo
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Camila C Portugal
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - João B Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| |
Collapse
|
5
|
Chronic oral exposure to short chain chlorinated paraffins induced testicular toxicity by promoting NRF2-mediated oxidative stress. Toxicol Lett 2023; 376:1-12. [PMID: 36642205 DOI: 10.1016/j.toxlet.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
As a widespread environmental contaminant, short chain chlorinated paraffins (SCCPs) has attracted great attention. However, the toxicity of SCCPs on male reproductive system remains ambiguous. In this study, we treated mice with SCCPs by gavage and investigated the toxic effects of SCCPs on testis. According to the results, the sperm parameters of mice were significantly reduced after exposure to 1, 10, 100 mg/kg body mass per day SCCPs for 35 days. SCCPs resulted in disorderly arranged seminiferous epithelium and increased apoptotic cells in testes. Both in vivo and in vitro experiments indicated that the oxidative stress was induced after SCCPs exposure, and dysfunction of nuclear factor erythroid-related factor (NRF2) signaling pathway played a role in this process. Moreover, resveratrol, an NRF2 activator, could alleviate the damage of SCCPs onmale reproductive system. Our study indicated that oxidative stress is the key point for explaining the testicular toxicity caused by SCCPs, and NRF2 could be used as a potential target for clinical treatment to alleviate the reproductive toxicity induced by SCCPs.
Collapse
|
6
|
Królicka E, Kieć-Kononowicz K, Łażewska D. Chalcones as Potential Ligands for the Treatment of Parkinson's Disease. Pharmaceuticals (Basel) 2022; 15:ph15070847. [PMID: 35890146 PMCID: PMC9317344 DOI: 10.3390/ph15070847] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/16/2022] Open
Abstract
Along with the increase in life expectancy, a significant increase of people suffering from neurodegenerative diseases (ND) has been noticed. The second most common ND, after Alzheimer’s disease, is Parkinson’s disease (PD), which manifests itself with a number of motor and non-motor symptoms that hinder the patient’s life. Current therapies can only alleviate those symptoms and slow down the progression of the disease, but not effectively cure it. So now, in addition to understanding the mechanism and causes of PD, it is also important to find a powerful way of treatment. It has been proved that in the etiology and course of PD, the essential roles are played by dopamine (DA) (an important neurotransmitter), enzymes regulating its level (e.g., COMT, MAO), and oxidative stress leading to neuroinflammation. Chalcones, due to their “simple” structure and valuable biological properties are considered as promising candidates for treatment of ND, also including PD. Here, we provide a comprehensive review of chalcones and related structures as potential new therapeutics for cure and prevention of PD. For this purpose, three databases (Pubmed, Scopus and Web of Science) were searched to collect articles published during the last 5 years (January 2018–February 2022). Chalcones have been described as promising enzyme inhibitors (MAO B, COMT, AChE), α-synuclein imaging probes, showing anti-neuroinflammatory activity (inhibition of iNOS or activation of Nrf2 signaling), as well as antagonists of adenosine A1 and/or A2A receptors. This review focused on the structure–activity relationships of these compounds to determine how a particular substituent or its position in the chalcone ring(s) (ring A and/or B) affects biological activity.
Collapse
|
7
|
Dong H, Wang L, Guo M, Stagos D, Giakountis A, Trachana V, Lin X, Liu Y, Liu M. Antioxidant and Anticancer Activities of Synthesized Methylated and Acetylated Derivatives of Natural Bromophenols. Antioxidants (Basel) 2022; 11:antiox11040786. [PMID: 35453471 PMCID: PMC9032154 DOI: 10.3390/antiox11040786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Natural bromophenols are important secondary metabolites in marine algae. Derivatives of these bromophenol are potential candidates for the drug development due to their biological activities, such as antioxidant, anticancer, anti-diabetic and anti-inflammatory activity. In our present study, we have designed and synthesized a series of new methylated and acetylated bromophenol derivatives from easily available materials using simple operation procedures and evaluated their antioxidant and anticancer activities on the cellular level. The results showed that 2.,3-dibromo-1-(((2-bromo-4,5-dimethoxybenzyl)oxy)methyl)-4,5-dimethoxybenzene (3b-9) and (oxybis(methylene))bis(4-bromo-6-methoxy-3,1-phenylene) diacetate (4b-3) compounds ameliorated H2O2-induced oxidative damage and ROS generation in HaCaT keratinocytes. Compounds 2.,3-dibromo-1-(((2-bromo-4,5-dimethoxybenzyl)oxy)methyl)-4,5-dimethoxybenzene (3b-9) and (oxybis(methylene) )bis(4-bromo-6-methoxy-3,1-phenylene) diacetate (4b-3) also increased the TrxR1 and HO-1 expression while not affecting Nrf2 expression in HaCaT. In addition, compounds (oxybis(methylene)bis(2-bromo-6-methoxy-4,1-phenylene) diacetate (4b-4) inhibited the viability and induced apoptosis of leukemia K562 cells while not affecting the cell cycle distribution. The present work indicated that some of these bromophenol derivatives possess significant antioxidant and anticancer potential, which merits further investigation.
Collapse
Affiliation(s)
- Hui Dong
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (H.D.); (L.W.); (M.G.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Li Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (H.D.); (L.W.); (M.G.)
| | - Meng Guo
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (H.D.); (L.W.); (M.G.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Dimitrios Stagos
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece; (D.S.); (A.G.)
| | - Antonis Giakountis
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece; (D.S.); (A.G.)
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41500 Larissa, Greece;
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, 319 Zhongshan Road, Jiangyang, Luzhou 646000, China;
| | - Yankai Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (H.D.); (L.W.); (M.G.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: (Y.L.); (M.L.); Tel.: +86-532-8203-1905 (Y.L.); +86-532-8203-1980 (M.L.)
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (H.D.); (L.W.); (M.G.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: (Y.L.); (M.L.); Tel.: +86-532-8203-1905 (Y.L.); +86-532-8203-1980 (M.L.)
| |
Collapse
|
8
|
Lee JA, Kwon YW, Kim HR, Shin N, Son HJ, Cheong CS, Kim DJ, Hwang O. A Novel Pyrazolo[3,4- d]pyrimidine Induces Heme Oxygenase-1 and Exerts Anti-Inflammatory and Neuroprotective Effects. Mol Cells 2022; 45:134-147. [PMID: 34887364 PMCID: PMC8926863 DOI: 10.14348/molcells.2021.0074] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/14/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
The anti-oxidant enzyme heme oxygenase-1 (HO-1) is known to exert anti-inflammatory effects. From a library of pyrazolo[3,4-d]pyrimidines, we identified a novel compound KKC080096 that upregulated HO-1 at the mRNA and protein levels in microglial BV-2 cells. KKC080096 exhibited anti-inflammatory effects via suppressing nitric oxide, interleukin-1β (IL-1β), and iNOS production in lipopolysaccharide (LPS)-challenged cells. It inhibited the phosphorylation of IKK and MAP kinases (p38, JNK, ERK), which trigger inflammatory signaling, and whose activities are inhibited by HO-1. Further, KKC080096 upregulated anti-inflammatory marker (Arg1, YM1, CD206, IL-10, transforming growth factor-β [TGF-β]) expression. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, KKC080096 lowered microglial activation, protected the nigral dopaminergic neurons, and nigral damage-associated motor deficits. Next, we elucidated the mechanisms by which KKC080096 upregulated HO-1. KKC080096 induced the phosphorylation of AMPK and its known upstream kinases LKB1 and CaMKKbeta, and pharmacological inhibition of AMPK activity reduced the effects of KKC080096 on HO-1 expression and LPS-induced NO generation, suggesting that KKC080096-induced HO-1 upregulation involves LKB1/AMPK and CaMKKbeta/AMPK pathway activation. Further, KKC080096 caused an increase in cellular Nrf2 level, bound to Keap1 (Nrf2 inhibitor protein) with high affinity, and blocked Keap1-Nrf2 interaction. This Nrf2 activation resulted in concurrent induction of HO-1 and other Nrf2-targeted antioxidant enzymes in BV-2 and in dopaminergic CATH.a cells. These results indicate that KKC080096 is a potential therapeutic for oxidative stress- and inflammation-related neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Ji Ae Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Young-Won Kwon
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hye Ri Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Nari Shin
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hyo Jin Son
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Chan Seong Cheong
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Dong Jin Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
9
|
Cao B, Zhang Y, Chen J, Wu P, Dong Y, Wang Y. Neuroprotective effects of liraglutide against inflammation through the AMPK/NF-κB pathway in a mouse model of Parkinson's disease. Metab Brain Dis 2022; 37:451-462. [PMID: 34817756 DOI: 10.1007/s11011-021-00879-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/15/2021] [Indexed: 03/12/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with increasing incidence in aged populations, second only to Alzheimer's disease. Increasing evidence has shown that inflammation plays an important role in the occurrence and development of Parkinson's disease. Growing evidence has shown that AMP-activated protein kinase (AMPK) and NF-κB are closely related to inflammation. Glucagon-like peptide 1 (GLP-1) is a hormone that is primarily secreted by intestinal endocrine L cells, and it has a variety of physiology through binding to GLP-1 receptor. GLP-1can be used for treatment of type 2 diabetes. In addition, GLP-1 also has anti-neuroinflammation activity. However, the exact mechanism behind how GLP-1 regulates neuroinflammation remains unclear. This study was designed to examine the effect of liraglutide on 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP)-induced injury in mice and its potential mechanism of action. Results showed that liraglutide dose-dependently ameliorated mouse behavior including swimming time and locomotor activity, increased the number of tyrosine hydroxylase (TH)-positive neurons and protein level, and reduced Iba1 and GFAP expression in the substantia nigra (SN). Liraglutide treatment also increased p-AMPK expression and reduced NF-κB protein level. Applying the AMPK inhibitor Dorsomorphin (Compound C) reversed the effect of liraglutide-reducing p-AMPK and increasing NF-κB expression. Finally, GFAP protein level increased, along with a decrease in TH expression. In conclusion, these results suggest that liraglutide can suppress neuroinflammation. Moreover, this effect is mediated through the AMPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bing Cao
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Yanqiu Zhang
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Jinhu Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Pengyue Wu
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Yuxuan Dong
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Yanqin Wang
- Department of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China.
| |
Collapse
|
10
|
Kang K, Xia A, Meng F, Chunyu J, Sun X, Ren G, Yu D, Jiang X, Tang L, Xiao W, Li D. FGF21 alleviates chronic inflammatory injury in the aging process through modulating polarization of macrophages. Int Immunopharmacol 2021; 96:107634. [PMID: 33872851 DOI: 10.1016/j.intimp.2021.107634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023]
Abstract
Previous studies reported that FGF21 prolongs life span and delays the body senescence, but the mechanism is not clear. The present study was designed to investigate the effects of FGF21 on hepatic senescence in aging mice and further research the mechanism. The 14-month-old male mice were administered with PBS, FGF21 or metformin once daily for 6 months. Results showed that FGF21 alleviated liver injury and inhibited accumulation of senescence markers SASP, P53 and P16 in the livers of aging mice. Subsequently we found that the aging mice treated by FGF21 showed transition of type 1 macrophages (M1) to type 2 macrophages (M2) in the livers. Next, we used THP-1 macrophages triggered by LPS to study effects of FGF21 on macrophages. Macrophages triggered by LPS exhibited features of M1, but the addition of FGF21 decreased the expression of M1 markers, and promoted the macrophages to exhibit features of M2. Results showed that the effects of FGF21 on macrophages were associated with the AMPK pathway. After adding AMPK inhibitor, the effects of FGF21 were inhibited, which was associated with the NF-κB signaling pathway. Finally, co-culturing differentiated macrophages and hepatocytes, we found that the large amount of pro-inflammatory factors such as IL-6 promoted hepatocyte senescence, which exhibited enhanced P53, P16 and β-galactosidase. This was contrary to hepatocytes co-cultured with macrophages treated by FGF21. These results indicate that FGF21 alleviates hepatic senescence injury by modulating the polarization of macrophages through the AMPK /NF-κB signaling pathway.
Collapse
Affiliation(s)
- Kai Kang
- Northeast Agricultural University, Harbin, China.
| | - Anran Xia
- Northeast Agricultural University, Harbin, China.
| | - Fanrui Meng
- Northeast Agricultural University, Harbin, China.
| | - Jian Chunyu
- Northeast Agricultural University, Harbin, China.
| | - Xu Sun
- Northeast Agricultural University, Harbin, China.
| | - Guiping Ren
- Northeast Agricultural University, Harbin, China.
| | - Dan Yu
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical CO. LTD, Lianyungang, Jiangsu, China.
| | | | - Lei Tang
- Harbin Weike Biotechnology CO. LTD, Harbin, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical CO. LTD, Lianyungang, Jiangsu, China.
| | - Deshan Li
- Northeast Agricultural University, Harbin, China.
| |
Collapse
|
11
|
Nikbakhtzadeh M, Shaerzadeh F, Ashabi G. Highlighting the protective or degenerative role of AMPK activators in dementia experimental models. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:786-801. [PMID: 34042039 DOI: 10.2174/1871527320666210526160214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/02/2020] [Accepted: 12/21/2020] [Indexed: 11/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is a serine/threonine kinase and a driving or deterrent factor in the development of neurodegenerative diseases and dementia. AMPK affects intracellular proteins like the mammalian target of rapamycin (mTOR). Peroxisome proliferator-activated receptor-γ coactivator 1-α (among others) contributes to a wide range of intracellular activities based on its downstream molecules such as energy balancing (ATP synthesis), extracellular inflammation, cell growth, and neuronal cell death (such as apoptosis, necrosis, and necroptosis). Several studies have looked at the dual role of AMPK in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington disease (HD) but the exact effect of this enzyme on dementia, stroke, and motor neuron dysfunction disorders has not been elucidated yet. In this article, we review current research on the effects of AMPK on the brain to give an overview of the relationship. More specifically, we review the neuroprotective or neurodegenerative effects of AMPK or AMPK activators like metformin, resveratrol, and 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside on neurological diseases and dementia, which exert through the intracellular molecules involved in neuronal survival or death.
Collapse
Affiliation(s)
- Marjan Nikbakhtzadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Shaerzadeh
- Department of Neuroscience, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, United States
| | - Ghorbangol Ashabi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Kang K, Xu P, Wang M, Chunyu J, Sun X, Ren G, Xiao W, Li D. FGF21 attenuates neurodegeneration through modulating neuroinflammation and oxidant-stress. Biomed Pharmacother 2020; 129:110439. [PMID: 32768941 DOI: 10.1016/j.biopha.2020.110439] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Previous studies indicate that FGF21 has ability to repair nerve injury, but the specific mechanism is less studied. The present study was designed to investigate the effects of FGF21 on neurodegeneration changes in aging and diabetic mice and its mechanism. The diabetic and aging mice were used to study the effects of FGF21 on neurodegeneration and possible mechanisms. These mice were administrated with PBS, FGF21 or metformin once daily for 4 or 6 months, then the mechanism was studied in SH-SY5Y cells. The relevant gene expression for neurodegeneration was assessed by Quantitative Real Time-PCR, Western blot, H&E staining, immunohistochemistry and ELISA. The Western blot results of NeuN showed that FGF21 inhibited the loss of neurons in diabetic and aging mice. H&E staining results showed that the karyopyknosis and tissue edema around dentate gyrus and Cornu Amonis 3 (CA3) area of hippocampus were also inhibited by FGF21 in aging and diabetes mice. In vivo results revealed that administration of FGF21 suppressed the aggregation of tau and β-amyloid1-42 in the brains of diabetic and aging mice. The aggregation resulted in apoptosis of neurons. Meanwhile, FGF21 significantly reduced the expression of Iba1, NF-κB, IL6 and IL8 (p < 0.05) and enhanced anti-oxidant enzymes (p < 0.05) in aging and diabetic mice. In addition, the phosphorylation of AKT and AMPKα were increased by FGF21 treatment. In vitro experiment showed that the aggregation of tau and β-amyloid1-42 wereincreased by LPS in SH-SY5Y cells, and FGF21 inhibited the aggregation through inhibiting the expression of NF-κB and promoting the phosphorylation of AKT and AMPKα. In conclusion, FGF21 attenuates neurodegeneration by reducing neuroinflammation and oxidant stress through regulating the NF-κB pathway and AMPKα/AKT pathway, which enhances the protective effect on mitochondria in neurons.
Collapse
Affiliation(s)
- Kai Kang
- Northeast Agricultural University, Harbin, China.
| | - Pengfei Xu
- National Laboratory of Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Fuwai Hospital, Beijing, China.
| | - Mengxia Wang
- Xinke College of Henan Institute of Science and Technology, China.
| | - Jian Chunyu
- Northeast Agricultural University, Harbin, China.
| | - Xu Sun
- Northeast Agricultural University, Harbin, China.
| | - Guiping Ren
- Northeast Agricultural University, Harbin, China.
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical CO. LTD, State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu, Lianyungang, China.
| | - Deshan Li
- Northeast Agricultural University, Harbin, China.
| |
Collapse
|
13
|
Lee JA, Kim HR, Son HJ, Shin N, Han SH, Cheong CS, Kim DJ, Hwang O. A novel pyrazolo [3,4-d] pyrimidine, KKC080106, activates the Nrf2 pathway and protects nigral dopaminergic neurons. Exp Neurol 2020; 332:113387. [PMID: 32580013 DOI: 10.1016/j.expneurol.2020.113387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/20/2020] [Accepted: 06/18/2020] [Indexed: 11/26/2022]
Abstract
The transcription factor nuclear factor-erythroid 2-related factor-2 (Nrf2) is known to induce neuroprotective and anti-inflammatory effects and is considered to be an excellent molecular target for drugs related to neurodegenerative disease therapy. Nrf2 activators previously tested in clinical trials were electrophilic, causing adverse effects due to non-selective and covalent modification of cellular thiols. In order to circumvent this issue, we constructed and screened a chemical library consisting of 241 pyrazolo [3,4-d] pyrimidine derivatives and discovered a novel, non-electrophilic compound: 1-benzyl-6-(methylthio)-N-(1-phenylethyl)-1H-pyrazolo[3,4-d]pyrimidine-4-amine (KKC080106). KKC080106 was able to activate Nrf2 signaling as it increases the cellular levels of Nrf2, binds to the Nrf2 inhibitor protein Keap1, and causes the accumulation of nuclear Nrf2. We also observed an increase in the expression levels of Nrf2-dependent genes for antioxidative/neuroprotective enzymes in dopaminergic neuronal cells. In addition, in lipopolysaccharide-activated microglia, KKC080106 suppressed the generation of the proinflammatory markers, such as IL-1β, TNF-α, cyclooxygenase-2, inducible nitric oxide synthase, and nitric oxide, and inhibited the phosphorylation of kinases known to be involved in inflammatory signaling, such as IκB kinase, p38, JNK, and ERK. As a drug, KKC080106 exhibited excellent stability against plasma enzymes and a good safety profile, evidenced by no mortality after the administration of 2000 mg/kg body weight, and minimal inhibition of the hERG channel activity. Pharmacokinetic analysis revealed that KKC080106 has good bioavailability and enters the brain after oral and intravenous administration, in both rats and mice. In MPTP-treated mice that received KKC080106 orally, the compound blocked microglial activation, protected the nigral dopaminergic neurons from degeneration, and prevented development of the dopamine deficiency-related motor deficits. These results suggest that KKC080106 has therapeutic potential for neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Ji Ae Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hye Ri Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo Jin Son
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Nari Shin
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Se Hee Han
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chan Seong Cheong
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Dong Jin Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea.
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
14
|
Chen D, Wu YX, Qiu YB, Wan BB, Liu G, Chen JL, Lu MD, Pang QF. Hyperoside suppresses hypoxia-induced A549 survival and proliferation through ferrous accumulation via AMPK/HO-1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 67:153138. [PMID: 31881478 DOI: 10.1016/j.phymed.2019.153138] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/01/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Hypoxia is commonly existed in tumors and lead to cancer cell chemo/radio-resistance. It is well-recognized that tumor hypoxia is a major challenge for the treatment of various solid tumors. Hyperoside (quercetin-3-O-galactoside, Hy) possesses antioxidant effects and has been reported to protect against hypoxia/reoxygenation induced injury in cardiomyocytes. Therefore, Hy may be attractive compound applicable to hypoxia-related diseases. PURPOSE This study was designed to determine the role of Hy in hypoxia-induced proliferation of non-small cell lung cancer cells and the underlying mechanism. STUDY DESIGN AND METHODS A549, a human non-small cell lung cancer (NSCLC) cell line, was used in the present study. 1% O2 was used to mimic the in vivo hypoxic condition of NSCLC. The potential mechanisms of Hy on hypoxia-induced A549 survival and proliferation, as well as the involvement of AMPK/HO-1 pathway were studied via CCK-8 assay, EdU staining, flow cytometry, qRT-PCR and western blot. RESULTS We showed that pretreatment with Hy suppressed hypoxia-induced A549 survival and proliferation in dose-dependent manner. In terms of mechanism, hypoxia-treated A549 showed the lower AMPK phosphorylation and the reduced HO-1 expression, which were reversed by Hy pretreatment. Both AMPK inhibitor (Compound C) and HO-1 activity inhibitor (Zinc protoporphyrin IX) abolished Hy-evoked A549 cell death under hypoxia stimuli. Of note, Ferrous iron contributed to Hy-induced A549 cell death under hypoxia, while Hy had no effect on lipid peroxidation under hypoxia. CONCLUSION Taken together, our results highlighted the beneficial role of Hy against hypoxia-induced A549 survival and proliferation through ferrous accumulation via AMPK/HO-1 axis.
Collapse
Affiliation(s)
- Dan Chen
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China
| | - Ya-Xian Wu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China
| | - Yu-Bao Qiu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China
| | - Bin-Bin Wan
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China
| | - Gang Liu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China
| | - Jun-Liang Chen
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China
| | - Mu-Dan Lu
- Central Laboratory, The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214122, Jiangsu Province, People's Republic of China.
| | - Qing-Feng Pang
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, Jiangsu Province, People's Republic of China.
| |
Collapse
|
15
|
Lee JA, Kim DJ, Hwang O. KMS99220 Exerts Anti-Inflammatory Effects, Activates the Nrf2 Signaling and Interferes with IKK, JNK and p38 MAPK via HO-1. Mol Cells 2019; 42:702-710. [PMID: 31656063 PMCID: PMC6821456 DOI: 10.14348/molcells.2019.0129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/10/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation is an important contributor to the pathogenesis of neurodegenerative disorders including Parkinson's disease (PD). We previously reported that our novel synthetic compound KMS99220 has a good pharmacokinetic profile, enters the brain, exerts neuroprotective effect, and inhibits NFκB activation. To further assess the utility of KMS99220 as a potential therapeutic agent for PD, we tested whether KMS99220 exerts an anti-inflammatory effect in vivo and examined the molecular mechanism mediating this phenomenon. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, oral administration of KMS99220 attenuated microglial activation and decreased the levels of inducible nitric oxide synthase and interleukin 1 beta (IL-1b) in the nigrostriatal system. In lipopolysaccharide (LPS)-challenged BV-2 microglial cells, KMS99220 suppressed the production and expression of IL-1b. In the activated microglia, KMS99220 reduced the phosphorylation of IκB kinase, c-Jun N-terminal kinase, and p38 MAP kinase; this effect was mediated by heme oxygenase-1 (HO-1), as both gene silencing and pharmacological inhibition of HO-1 abolished the effect of KMS99220. KMS99220 induced nuclear translocation of the transcription factor Nrf2 and expression of the Nrf2 target genes including HO-1. Together with our earlier findings, our current results show that KMS99220 may be a potential therapeutic agent for neuroinflammation-related neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
- Ji Ae Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Dong Jin Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792,
Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505,
Korea
| |
Collapse
|