1
|
Lyu J, Zhang H, Wang C, Pan M. New insight in treating autoimmune diseases by targeting autophagy. Autoimmunity 2024; 57:2351872. [PMID: 38739691 DOI: 10.1080/08916934.2024.2351872] [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: 01/22/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Autophagy is a highly conserved biological process in eukaryotes, which degrades cellular misfolded proteins, damaged organelles and invasive pathogens in the lysosome-dependent manner. Autoimmune diseases caused by genetic elements, environments and aberrant immune responses severely impact patients' living quality and even threaten life. Recently, numerous studies have reported autophagy can regulate immune responses, and play an important role in autoimmune diseases. In this review, we summarised the features of autophagy and autophagy-related genes, enumerated some autophagy-related genes involved in autoimmune diseases, and further overviewed how to treat autoimmune diseases through targeting autophagy. Finally, we outlooked the prospect of relieving and curing autoimmune diseases by targeting autophagy pathway.
Collapse
Affiliation(s)
- Jiao Lyu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hongqian Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chaoyang Wang
- The Key Medical Laboratory for Chemical Poison Detection of Henan Province, The Third People's Hospital of Henan Province, Zhengzhou, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China
| | - Mingyu Pan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Xue Y, Zhang L, Chu L, Song Z, Zhang B, Su X, Liu W, Li X. JAK2/STAT3 Pathway Inhibition by AG490 Ameliorates Experimental Autoimmune Encephalomyelitis via Regulation of Th17 Cells and Autophagy. Neuroscience 2024; 552:65-75. [PMID: 38885894 DOI: 10.1016/j.neuroscience.2024.06.009] [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: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory condition affecting the central nervous system, and experimental autoimmune encephalomyelitis (EAE) animal models have been extensively used to study it. T-helper 17 cells, which produce interleukin-17(IL-17), play crucial roles in MS pathogenesis, and the JAK2/STAT3 pathway has an essential function in their differentiation from naive CD4 + T cells. This study investigated the effects of the JAK2/STAT3 pathway inhibitor AG490 on EAE in vivo and in vitro, as well as the underlying mechanisms. AG490 ameliorated EAE severity and attenuated its typical symptoms by downregulating proteins associated with the JAK2/STAT3 pathway. Furthermore, it decreased T-helper 17 cell differentiation from naive CD4 + T cells by inactivating STAT3. In addition, it conferred protective effects against EAE by restoring autophagy. These findings indicate the potential of AG490 as a candidate anti-MS therapeutic.
Collapse
Affiliation(s)
- Yumei Xue
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Lu Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lifang Chu
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhe Song
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Bing Zhang
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaohui Su
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Wanhu Liu
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaobing Li
- Department of Pharmacy, Shijiazhuang People's Hospital, Shijiazhuang, China.
| |
Collapse
|
3
|
Ren W, Sun Y, Zhao L, Shi X. NLRP3 inflammasome and its role in autoimmune diseases: A promising therapeutic target. Biomed Pharmacother 2024; 175:116679. [PMID: 38701567 DOI: 10.1016/j.biopha.2024.116679] [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: 02/16/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
The NOD-like receptor protein 3 (NLRP3) inflammasome is a protein complex that regulates innate immune responses by activating caspase-1 and the inflammatory cytokines IL-1β and IL-18. Numerous studies have highlighted its crucial role in the pathogenesis and development of inflammatory bowel disease, rheumatoid arthritis, systemic lupus erythematosus, autoimmune thyroid diseases, and other autoimmune diseases. Therefore, investigating the underlying mechanisms of NLRP3 in disease and targeted drug therapies holds clinical significance. This review summarizes the structure, assembly, and activation mechanisms of the NLRP3 inflammasome, focusing on its role and involvement in various autoimmune diseases. This review also identifies studies where the involvement of the NLRP3 inflammasome in the disease mechanism within the same disease appears contradictory, as well as differences in NLRP3-related gene polymorphisms among different ethnic groups. Additionally, the latest therapeutic advances in targeting the NLRP3 inflammasome for autoimmune diseases are outlined, and novel clinical perspectives are discussed. Conclusively, this review provides a consolidated source of information on the NLRP3 inflammasome and may guide future research efforts that have the potential to positively impact patient outcomes.
Collapse
Affiliation(s)
- Wenxuan Ren
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Ying Sun
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Zhao
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Xiaoguang Shi
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
4
|
Wang X, Qu Y, Fan J, Ren H. Serum NfL and EGFR/NfL ratio mRNAs as biomarkers for phenotype and disease severity of myelin oligodendrocyte glycoprotein IgG-associated disease. Front Immunol 2024; 15:1388734. [PMID: 38807603 PMCID: PMC11130348 DOI: 10.3389/fimmu.2024.1388734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
Background and purpose Myelin oligodendrocyte glycoprotein (MOG) IgG is frequently elevated in pediatric patients with acquired demyelinating syndrome (ADS). However, no specific biomarkers exist for phenotype classification, symptom severity, prognosis, and treatment guidance of MOG-IgG-associated disease (MOGAD). This study evaluated neurofilament light chain (NfL) and endothelial growth factor receptor (EGFR) mRNA expression levels in serum and cerebrospinal fluid (CSF) as potential biomarkers for MOGAD in Chinese children. Methods This was a cross-sectional and single-center study. We enrolled 22 consecutive pediatric patients hospitalized with MOGAD and 20 control pediatric patients hospitalized for noninflammatory neurological diseases in Hebei Children's Hospital. Serum and CSF were collected from MOGAD patients within 3 days before immunotherapy. The mRNA levels of NfL and EGFR in serum and CSF were measured by real-time polymerase chain reaction (qPCR), and the EGFR/NfL ratio mRNA was calculated. These measurement values were then compared between disease groups and among MOGAD phenotypes. In addition, the correlations between the mRNAs of three markers (NfL, EGFR, EGFR/NfL ratio), extended disability status scale (EDSS) scores, and clinical phenotypes were analyzed. Results Serum and CSF NfL mRNA levels were significantly higher of acute-stage MOGAD patients than those of control patients (p< 0.05 and p< 0.01, respectively), while the mRNA levels of serum EGFR and EGFR/NfL ratio were significantly lower of MOGAD patients than those of controls (p < 0.05, p < 0.0001). Serum NfL mRNA was significantly correlated with mRNA of serum EGFR (r =0.480, p < 0.05). Serum and CSF NfL mRNA levels in MOGAD patients with the ADEM-like phenotype were also significantly higher than those in control patients (p < 0.01, p < 0.01) and optic neuritis (ON) phenotype (p < 0.05, p < 0.05). Both mRNAs of NfL in CSF and EGFR/NfL ratio in serum were correlated with EDSS scores (p < 0.05, r = 0.424; p < 0.05, r= -0.521). Conclusion The mRNA levels of elevated NfL in serum and CSF as well as lower EGFR and EGFR/NfL ratio in serum could help distinguish acute-phase MOGAD. Higher mRNA levels of NfL in serum and CSF of MOGAD patients help distinguish ADEM-like phenotype. In addition, serum EGFR/NfL mRNA ratio is indicative of disease severity in pediatric patients with MOGAD. Further investigations are warranted to elucidate the pathological mechanisms underlying these associations.
Collapse
Affiliation(s)
- Xin Wang
- Second Department of Neurology, Hebei Children’s Hospital, Shijiazhuang, China
| | - Yi Qu
- Department of Science and Education, Hebei Children’s Hospital, Shijiazhuang, China
| | - Jiayu Fan
- Second Department of Neurology, Hebei Children’s Hospital, Shijiazhuang, China
| | - Huiqiang Ren
- Department of Pathology, Hebei Children’s Hospital, Shijiazhuang, China
| |
Collapse
|
5
|
Wu J, Han K, Sack MN. Targeting NAD+ Metabolism to Modulate Autoimmunity and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1043-1050. [PMID: 38498807 PMCID: PMC10954088 DOI: 10.4049/jimmunol.2300693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/18/2023] [Indexed: 03/20/2024]
Abstract
NAD+ biology is involved in controlling redox balance, functioning as a coenzyme in numerous enzymatic reactions, and is a cofactor for Sirtuin enzymes and a substrate for multiple regulatory enzyme reactions within and outside the cell. At the same time, NAD+ levels are diminished with aging and are consumed during the development of inflammatory and autoimmune diseases linked to aberrant immune activation. Direct NAD+ augmentation via the NAD+ salvage and Priess-Handler pathways is being investigated as a putative therapeutic intervention to improve the healthspan in inflammation-linked diseases. In this review, we survey NAD+ biology and its pivotal roles in the regulation of immunity and inflammation. Furthermore, we discuss emerging studies evaluate NAD+ boosting in murine models and in human diseases, and we highlight areas of research that remain unresolved in understanding the mechanisms of action of these nutritional supplementation strategies.
Collapse
Affiliation(s)
- Jing Wu
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
6
|
Zeng X, Zhang K, Liang M, Yu B, Zhang P, Mehmood A, Zhang H. NAD + affects differentially expressed genes- MBOAT2- SLC25A21- SOX6 in experimental autoimmune encephalomyelitis model. Int J Neurosci 2024:1-8. [PMID: 38315116 DOI: 10.1080/00207454.2024.2313022] [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: 01/23/2024] [Accepted: 01/28/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Nicotinamide adenine dinucleotide (NAD+) plays a key role in neuroinflammation and neurodegeneration and provides anti-inflammatory and neuroprotective effects in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). AIM In this study, we aimed to investigate whether NAD+ affects differentially expressed genes (DEGs) in splenocytes of EAE mice to reveal candidate genes for the pathogenesis of MS. METHODS The EAE model was used to perform an intervention on NAD+ to investigate its potential as a protective agent in inflammation and demyelination. Transcriptome analysis of nerve tissue was carried out to gain better insights into NAD+ function. Effects of NAD+ on DEGs in the splenocytes of EAE mice were investigated to determine its anti-inflammatory effect. RESULTS NAD+ in EAE mice showed the clinical score was significantly improved (EAE 3.190 ± 0.473 vs. NAD+ 2.049 ± 0.715). DEGs (MBOAT2, SLC25A21, and SOX6) between the EAE and the EAE + NAD+ groups showed that SOX6 was significantly improved after NAD+ treatment compared with the EAE group, and other indicators were improved but did not reach statistical significance. NAD+ exhibited clinical scores in EAE mice, and key inflammation was ameliorated in EAE mice spleen after NAD+ intervention, while transcriptome analysis between EAE and EAE + NAD+ groups showed several DEGs in the underlying mechanism. CONCLUSION NAD+ on DEGs attenuates disease severity in EAE. Transcriptome analysis on nerve tissue reveals several protein targets in the underlying mechanisms. However, NAD+ does not significantly improve DEGs in the splenocytes of the EAE model.
Collapse
Affiliation(s)
- Xu Zeng
- Department of Neurosurgery, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Kexue Zhang
- Senior Department of Pediatric, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Ming Liang
- Senior Department of Pediatric, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Bin Yu
- Senior Department of Pediatric, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Peng Zhang
- Department of Neurosurgery, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Hongtian Zhang
- Department of Neurosurgery, The Seventh Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Ravichandran KA, Heneka MT. Inflammasomes in neurological disorders - mechanisms and therapeutic potential. Nat Rev Neurol 2024; 20:67-83. [PMID: 38195712 DOI: 10.1038/s41582-023-00915-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/11/2024]
Abstract
Inflammasomes are molecular scaffolds that are activated by damage-associated and pathogen-associated molecular patterns and form a key element of innate immune responses. Consequently, the involvement of inflammasomes in several diseases that are characterized by inflammatory processes, such as multiple sclerosis, is widely appreciated. However, many other neurological conditions, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, stroke, epilepsy, traumatic brain injury, sepsis-associated encephalopathy and neurological sequelae of COVID-19, all involve persistent inflammation in the brain, and increasing evidence suggests that inflammasome activation contributes to disease progression in these conditions. Understanding the biology and mechanisms of inflammasome activation is, therefore, crucial for the development of inflammasome-targeted therapies for neurological conditions. In this Review, we present the current evidence for and understanding of inflammasome activation in neurological diseases and discuss current and potential interventional strategies that target inflammasome activation to mitigate its pathological consequences.
Collapse
Affiliation(s)
- Kishore Aravind Ravichandran
- Department of Neuroinflammation, Institute of innate immunity, University of Bonn Medical Center Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Esch-sur-Alzette, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, MA, USA.
| |
Collapse
|
8
|
Al-kuraishy HM, Jabir MS, Al-Gareeb AI, Saad HM, Batiha GES, Klionsky DJ. The beneficial role of autophagy in multiple sclerosis: Yes or No? Autophagy 2024; 20:259-274. [PMID: 37712858 PMCID: PMC10813579 DOI: 10.1080/15548627.2023.2259281] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/16/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic progressive demyelinating disease of the central nervous system (CNS) due to an increase of abnormal peripherally auto-reactive T lymphocytes which elicit autoimmunity. The main pathophysiology of MS is myelin sheath damage by immune cells and a defect in the generation of myelin by oligodendrocytes. Macroautophagy/autophagy is a critical degradation process that eliminates dysfunctional or superfluous cellular components. Autophagy has the property of a double-edged sword in MS in that it may have both beneficial and detrimental effects on MS neuropathology. Therefore, this review illustrates the protective and harmful effects of autophagy with regard to this disease. Autophagy prevents the progression of MS by reducing oxidative stress and inflammatory disorders. In contrast, over-activated autophagy is associated with the progression of MS neuropathology and in this case the use of autophagy inhibitors may alleviate the pathogenesis of MS. Furthermore, autophagy provokes the activation of different immune and supporting cells that play an intricate role in the pathogenesis of MS. Autophagy functions in the modulation of MS neuropathology by regulating cell proliferation related to demyelination and remyelination. Autophagy enhances remyelination by increasing the activity of oligodendrocytes, and astrocytes. However, autophagy induces demyelination by activating microglia and T cells. In conclusion, specific autophagic activators of oligodendrocytes, and astrocytes, and specific autophagic inhibitors of dendritic cells (DCs), microglia and T cells induce protective effects against the pathogenesis of MS.Abbreviations: ALS: amyotrophic lateral sclerosis; APCs: antigen-presenting cells; BBB: blood-brain barrier; CSF: cerebrospinal fluid; CNS: central nervous system; DCs: dendritic cells; EAE: experimental autoimmune encephalomyelitis; ER: endoplasmic reticulum; LAP: LC3-associated phagocytosis; MS: multiple sclerosis; NCA: non-canonical autophagy; OCBs: oligoclonal bands; PBMCs: peripheral blood mononuclear cells; PD: Parkinson disease; ROS: reactive oxygen species; UPR: unfolded protein response.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Iraq, Baghdad
| | - Majid S. Jabir
- Department of Applied Science, University of Technology, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Iraq, Baghdad
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, El Beheira, Egypt
| | | |
Collapse
|
9
|
Kitaoka Y, Sase K. Molecular aspects of optic nerve autophagy in glaucoma. Mol Aspects Med 2023; 94:101217. [PMID: 37839231 DOI: 10.1016/j.mam.2023.101217] [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: 06/30/2023] [Revised: 09/24/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
The optic nerve consists of the glia, vessels, and axons including myelin and axoplasm. Since axonal degeneration precedes retinal ganglion cell death in glaucoma, the preceding axonal degeneration model may be helpful for understanding the molecular mechanisms of optic nerve degeneration. Optic nerve samples from these models can provide information on several aspects of autophagy. Autophagosomes, the most typical organelles expressing autophagy, are found much more frequently inside axons than around the glia. Thus, immunoblot findings from the optic nerve can reflect the autophagy state in axons. Autophagic flux impairment may occur in degenerating optic nerve axons, as in other central nervous system neurodegenerative diseases. Several molecular candidates are involved in autophagy enhancement, leading to axonal protection. This concept is an attractive approach to the prevention of further retinal ganglion cell death. In this review, we describe the factors affecting autophagy, including nicotinamide riboside, p38, ULK, AMPK, ROCK, and SIRT1, in the optic nerve and propose potential methods of axonal protection via enhancement of autophagy.
Collapse
Affiliation(s)
- Yasushi Kitaoka
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan; Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Kana Sase
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
10
|
Kaffe D, Kaplanis SI, Karagogeos D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Curr Issues Mol Biol 2023; 45:9526-9548. [PMID: 38132442 PMCID: PMC10742427 DOI: 10.3390/cimb45120596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The dysfunction of myelinating glial cells, the oligodendrocytes, within the central nervous system (CNS) can result in the disruption of myelin, the lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. This leads to axonal degeneration and motor/cognitive impairments. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed oligodendrocytes. Apart from oligodendrocytes, the two other main glial cell types of the CNS, microglia and astrocytes, play a pivotal role in remyelination. Following a demyelination insult, microglia can phagocytose myelin debris, thus permitting remyelination, while the developing neuroinflammation in the demyelinated region triggers the activation of astrocytes. Modulating the profile of glial cells can enhance the likelihood of successful remyelination. In this context, recent studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. In this Review, we examine the role of substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. Such substances, called caloric restriction mimetics, have been shown to decelerate the aging process by mitigating age-related ailments, with their mechanisms of action intricately linked to the induction of autophagic processes.
Collapse
Affiliation(s)
- Despoina Kaffe
- Department of Biology, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
| | - Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
11
|
Mehmood A, Song S, Du X, Yan H, Wang X, Guo L, Li B. mRNA expression profile reveals differentially expressed genes in splenocytes of experimental autoimmune encephalomyelitis model. Int J Exp Pathol 2023; 104:247-257. [PMID: 37427716 PMCID: PMC10500171 DOI: 10.1111/iep.12488] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/04/2023] [Accepted: 06/18/2023] [Indexed: 07/11/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a mouse model that can be used to investigate aetiology, pathogenesis, and treatment approaches for multiple sclerosis (MS). A novel integrated bioinformatics approach was used to understand the involvement of differentially expressed genes (DEGs) in the spleen of EAE mice through data mining of existing microarray and RNA-seq datasets. We screened differentially expressed mRNAs using mRNA expression profile data of EAE spleens taken from Gene Expression Omnibus (GEO). Functional and pathway enrichment analyses of DEGs were performed by Database for Annotation, Visualization, and Integrated Discovery (DAVID). Subsequently, the DEGs-encoded protein-protein interaction (PPI) network was constructed. The 784 DEGs in GSE99300 A.SW PP-EAE mice spleen mRNA profiles, 859 DEGs in GSE151701 EAE mice spleen mRNA profiles, and 646 DEGs in GSE99300 SJL/J PP-EAE mice spleen mRNA profiles were explored. Functional enrichment of 55 common DEGs among 3 sub-datasets revealed several immune-related terms, such as neutrophil extravasation, leucocyte migration, antimicrobial humoral immune response mediated by an antimicrobial peptide, toll-like receptor 4 bindings, IL-17 signalling pathway, and TGF-beta signalling pathway. In the screening of 10 hub genes, including MPO, ELANE, CTSG, LTF, LCN2, SELP, CAMP, S100A9, ITGA2B, and PRTN3, and in choosing and validating the 5 DEGs, including ANK1, MBOAT2, SLC25A21, SLC43A1, and SOX6, the results showed that SLC43A1 and SOX6 were significantly decreased in EAE mice spleen. Thus this study offers a list of genes expressed in the spleen that might play a key role in the pathogenesis of EAE.
Collapse
Affiliation(s)
- Arshad Mehmood
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Shuang Song
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Xiaochen Du
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Hongjing Yan
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Xuan Wang
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Li Guo
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| | - Bin Li
- Department of NeurologyThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurology of Hebei ProvinceShijiazhuangHebeiChina
| |
Collapse
|
12
|
Kaplanis SI, Kaffe D, Ktena N, Lygeraki A, Kolliniati O, Savvaki M, Karagogeos D. Nicotinamide enhances myelin production after demyelination through reduction of astrogliosis and microgliosis. Front Cell Neurosci 2023; 17:1201317. [PMID: 37663127 PMCID: PMC10469866 DOI: 10.3389/fncel.2023.1201317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Caloric restriction is the chronic reduction of total caloric intake without malnutrition and has attracted a lot of attention as, among multiple other effects, it attenuates demyelination and stimulates remyelination. In this study we have evaluated the effect of nicotinamide (NAM), a well-known caloric restriction mimetic, on myelin production upon demyelinating conditions. NAM is the derivative of nicotinic acid (vitamin B3) and a precursor of nicotinamide adenine dinucleotide (NAD+), a ubiquitous metabolic cofactor. Here, we use cortical slices ex vivo subjected to demyelination or cultured upon normal conditions, a lysolecithin (LPC)-induced focal demyelination mouse model as well as primary glial cultures. Our data show that NAM enhances both myelination and remyelination ex vivo, while it also induces myelin production after LPC-induced focal demyelination ex vivo and in vivo. The increased myelin production is accompanied by reduction in both astrogliosis and microgliosis in vivo. There is no direct effect of NAM on the oligodendrocyte lineage, as no differences are observed in oligodendrocyte precursor cell proliferation or differentiation or in the number of mature oligodendrocytes. On the other hand, NAM affects both microglia and astrocytes as it decreases the population of M1-activated microglia, while reducing the pro-inflammatory phenotype of astrocytes as assayed by the reduction of TNF-α. Overall, we show that the increased myelin production that follows NAM treatment in vivo is accompanied by a decrease in both astrocyte and microglia accumulation at the lesion site. Our data indicate that NAM influences astrocytes and microglia directly, in favor of the remyelination process by promoting a less inflammatory environment.
Collapse
Affiliation(s)
- Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Despoina Kaffe
- Department of Biology, University of Crete, Heraklion, Greece
| | - Niki Ktena
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | | | - Ourania Kolliniati
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece
- Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Maria Savvaki
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| |
Collapse
|
13
|
Panda C, Mahapatra RK. Bi-Directional Relationship Between Autophagy and Inflammasomes in Neurodegenerative Disorders. Cell Mol Neurobiol 2023; 43:115-137. [PMID: 35066716 DOI: 10.1007/s10571-021-01184-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/12/2021] [Indexed: 01/18/2023]
Abstract
The innate immune system, as the first line of cellular defense, triggers a protective response called inflammation when encountered with invading pathogens. Inflammasome is a multi-protein cytosolic signaling complex that induces inflammation and is critical for inflammation-induced pyroptotic cell death. Inflammasome activation has been found associated with neurodegenerative disorders (NDs), inflammatory diseases, and cancer. Autophagy is a crucial intracellular quality control and homeostasis process which removes the dysfunctional organelles, damaged proteins, and pathogens by sequestering the cytosolic components in a double-membrane vesicle, which eventually fuses with lysosome resulting in cargo degradation. Autophagy disruption has been observed in many NDs presented with persistent neuroinflammation and excessive inflammasome activation. An interplay between inflammation activation and the autophagy process has been realized over the last decade. In the case of NDs, autophagy regulates neuroinflammation load and cellular damage either by engulfing the misfolded protein deposits, dysfunctional mitochondria, or the inflammasome complex itself. A healthy two-way regulation between both cellular processes has been realized for cell survival and cell defense during inflammatory conditions. Therefore, clinical interest in the modulation of inflammasome activation by autophagy inducers is rapidly growing. In this review, we discuss the structural basis of inflammasome activation and the mechanistic ideas of the autophagy process in NDs. Along with comments on multiple ways of neuroinflammation regulation by microglial autophagy, we also present a perspective on pharmacological opportunities in this molecular interplay pertaining to NDs.
Collapse
Affiliation(s)
- Chinmaya Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India
| | - Rajani Kanta Mahapatra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
14
|
Ben-Zvi H, Rabinski T, Ofir R, Cohen S, Vatine GD. PLEKHM2 Loss of Function Impairs the Activity of iPSC-Derived Neurons via Regulation of Autophagic Flux. Int J Mol Sci 2022; 23:ijms232416092. [PMID: 36555735 PMCID: PMC9782635 DOI: 10.3390/ijms232416092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Pleckstrin Homology And RUN Domain Containing M2 (PLEKHM2) [delAG] mutation causes dilated cardiomyopathy with left ventricular non-compaction (DCM-LVNC), resulting in a premature death of PLEKHM2[delAG] individuals due to heart failure. PLEKHM2 is a factor involved in autophagy, a master regulator of cellular homeostasis, decomposing pathogens, proteins and other cellular components. Autophagy is mainly carried out by the lysosome, containing degradation enzymes, and by the autophagosome, which engulfs substances marked for decomposition. PLEKHM2 promotes lysosomal movement toward the cell periphery. Autophagic dysregulation is associated with neurodegenerative diseases' pathogenesis. Thus, modulation of autophagy holds considerable potential as a therapeutic target for such disorders. We hypothesized that PLEKHM2 is involved in neuronal development and function, and that mutated PLEKHM2 (PLEKHM2[delAG]) neurons will present impaired functions. Here, we studied PLEKHM2-related abnormalities in induced pluripotent stem cell (iPSC)-derived motor neurons (iMNs) as a neuronal model. PLEKHM2[delAG] iMN cultures had healthy control-like differentiation potential but exhibited reduced autophagic activity. Electrophysiological measurements revealed that PLEKHM2[delAG] iMN cultures displayed delayed functional maturation and more frequent and unsynchronized activity. This was associated with increased size and a more perinuclear lysosome cellular distribution. Thus, our results suggest that PLEKHM2 is involved in the functional development of neurons through the regulation of autophagic flux.
Collapse
Affiliation(s)
- Hadas Ben-Zvi
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Tatiana Rabinski
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Rivka Ofir
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Dead Sea & Arava Science Center, Masada 8691000, Israel
| | - Smadar Cohen
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Correspondence: (S.C.); (G.D.V.)
| | - Gad D. Vatine
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- The Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- The Zelman School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Correspondence: (S.C.); (G.D.V.)
| |
Collapse
|
15
|
Melatonin ameliorates disease severity in a mouse model of multiple sclerosis by modulating the kynurenine pathway. Sci Rep 2022; 12:15963. [PMID: 36153399 PMCID: PMC9509376 DOI: 10.1038/s41598-022-20164-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractMelatonin (MT), a neurohormone with immunomodulatory properties, is one of the metabolites produced in the brain from tryptophan (TRP) that has already strong links with the neuropathogenesis of Multiple sclerosis (MS). However, the exact molecular mechanisms behind that are not fully understood. There is some evidence showing that MS and MT are interconnected via different pathways: Relapses of MS has a direct correlation with a low level of MT secretion and a growing body of evidence suggest that MT be therapeutic in Experimental Autoimmune Encephalomyelitis (EAE, a recognise animal model of MS) severity. Previous studies have demonstrated that the kynurenine pathway (KP), the main pathway of TRP catabolism, plays a key role in the pathogenesis of MS in humans and in EAE. The present study aimed to investigate whether MT can improve clinical signs in the EAE model by modulating the KP. C57BL/6 mice were induced with EAE and received different doses of MT. Then the onset and severity of EAE clinical symptoms were recorded. Two biological factors, aryl hydrocarbon receptor (AhR) and NAD+ which closely interact in the KP were also assessed. The results indicated that MT treatment at all tested doses significantly decrease the EAE clinical scores and the number of demyelinating plaques. Furthermore, MT treatment reduced the mRNA expression of the KP regulatory enzyme indoleamine 2,3-dioxygenase 1(IDO-1) and other KP enzymes. We also found that MT treatment reduces the mRNA expression of the AhR and inhibits the enzyme Nicotinamide N-Methyltransferase (Nnmt) overexpression leading to an increase in NAD+ levels. Collectively, this study suggests that MT treatment may significantly attenuates the severity of EAE by altering the KP, AhR and NAD+ metabolism.
Collapse
|
16
|
Ding W, Cai C, Zhu X, Wang J, Jiang Q. Parthenolide ameliorates neurological deficits and neuroinflammation in mice with traumatic brain injury by suppressing STAT3/NF-κB and inflammasome activation. Int Immunopharmacol 2022; 108:108913. [PMID: 35729839 DOI: 10.1016/j.intimp.2022.108913] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/15/2022] [Accepted: 05/27/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) triggers a set of complex inflammation that results in secondary injury. Parthenolide (PTN) is a sesquiterpene lactone extracted from the herb Tanacetum parthenium (Feverfew) and has potent anti-inflammatory, anti-apoptosis and anti-oxidative stress effects in the central nervous system (CNS)-related diseases. This study focuses on investigating the potential neuroprotective effect of PTN on TBI and the related mechanism. METHODS Bv2 microglia, primary microglia were stimulated by LPS, and HT22 neuron cells were stimulated by OGD/R, and they were treated with different doses of PTN. The expression profiles of pro-inflammatory cytokines, proteins, oxidative stress mediators, STAT3/NF-κB pathway, inflammasomes were detected. Forty male/female C57BL/6 mice were randomly divided into the sham, PTN, TBI, and TBI + PTN groups (10 mice per group). A mouse TBI model was set up with a controlled cortical impact (CCI) device. The modified nerve severity score (mNSS) was implemented to check short-term neurological impairment in mice, and the mice's memory and learning were assessed by the Morris water maze test. The water content in the mice's brains was measured by the dry-wet method. Hematoxylin-eosin (H&E) staining, Nissl staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay were applied for neuronal apoptosis. RESULTS PTN dramatically alleviated LPS-induced inflammation in microglia, and OGD-mediated neuronal apoptosis and oxidative stress. In addition, PTN repressed LPS- or OGD-modulated STAT3/NF-κB and NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLR family CARD domain containing 4 (NLRC4) inflammasomes activation. Administering the STAT3 inhibitor Stattic or NF-κB inhibitor Bay 11-7082 attenuated PTN-mediated effects. In vivo, PTN treatment relieved neural function deficits, brain edema and neuron apoptosis and improved the memory and learning function of TBI mice. Additionally, PTN impeded microglial activation and reduced the production of pro-inflammatory cytokines in brain lesions of TBI mice. Furthermore, PTN hindered STAT3/NF-κB and inflammasome activation. CONCLUSION PTN can curb microglial activation and neuron apoptosis by dampening the STAT3/NF-κB pathway, thus exerting neuroprotective effects in TBI mice.
Collapse
Affiliation(s)
- Wei Ding
- Department of Neurosurgery, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430000, China; Department of Neurosurgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chen Cai
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaomin Zhu
- Department of Neurology, Guangxi University of Chinese Medicine, Nanning 530200 Guangxi, China
| | - Jing Wang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
17
|
Yu M, Zheng X, Cheng F, Shao B, Zhuge Q, Jin K. Metformin, Rapamycin, or Nicotinamide Mononucleotide Pretreatment Attenuate Cognitive Impairment After Cerebral Hypoperfusion by Inhibiting Microglial Phagocytosis. Front Neurol 2022; 13:903565. [PMID: 35769369 PMCID: PMC9234123 DOI: 10.3389/fneur.2022.903565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/10/2022] [Indexed: 12/02/2022] Open
Abstract
Vascular cognitive impairment (VCI) is the second leading form of dementia after Alzheimer's disease (AD) plaguing the elder population. Despite the enormous prevalence of VCI, the biological basis of this disease has been much less well-studied than that of AD, with no specific therapy currently existing to prevent or treat VCI. As VCI mainly occurs in the elderly, the role of anti-aging drugs including metformin, rapamycin, and nicotinamide mono nucleotide (NMN), and the underlying mechanism remain uncertain. Here, we examined the role of metformin, rapamycin, and NMN in cognitive function, white matter integrity, microglial response, and phagocytosis in a rat model of VCI by bilateral common carotid artery occlusion (BCCAO). BCCAO-induced chronic cerebral hypoperfusion could cause spatial working memory deficits and white matter lesions (WMLs), along with increasing microglial activation and phagocytosis compared to sham-operated rats. We found the cognitive impairment was significantly improved in BCCAO rats pretreated with these three drugs for 14 days before BCCAO compared with the vehicle group by the analysis of the Morris water maze and new object recognition tests. Pretreatment of metformin, rapamycin, or NMN also increased myelin basic protein (MBP, a marker for myelin) expression and reduced SMI32 (a marker for demyelinated axons) intensity and SMI32/MBP ratio compared with the vehicle group, suggesting that these drugs could ameliorate BCCAO-induced WMLs. The findings were confirmed by Luxol fast blue (LFB) stain, which is designed for staining myelin/myelinated axons. We further found that pretreatment of metformin, rapamycin, or NMN reduced microglial activation and the number of M1 microglia, but increased the number of M2 microglia compared to the vehicle group. Importantly, the number of MBP+/Iba1+/CD68+ microglia was significantly reduced in the BCCAO rats pretreated with these three drugs compared with the vehicle group, suggesting that these drugs suppress microglial phagocytosis. No significant difference was found between the groups pretreated with metformin, rapamycin, or NMN. Our data suggest that metformin, rapamycin, or NMN could protect or attenuate cognitive impairment and WMLs by modifying microglial polarization and inhibiting phagocytosis. The findings may open a new avenue for VCI treatment.
Collapse
Affiliation(s)
- Mengdi Yu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoying Zheng
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Fangyu Cheng
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Bei Shao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
- *Correspondence: Qichuan Zhuge
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- Kunlin Jin
| |
Collapse
|
18
|
Qian ZY, Kong RY, Zhang S, Wang BY, Chang J, Cao J, Wu CQ, Huang ZY, Duan A, Li HJ, Yang L, Cao XJ. Ruxolitinib attenuates secondary injury after traumatic spinal cord injury. Neural Regen Res 2022; 17:2029-2035. [PMID: 35142693 PMCID: PMC8848590 DOI: 10.4103/1673-5374.335165] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Excessive inflammation post-traumatic spinal cord injury (SCI) induces microglial activation, which leads to prolonged neurological dysfunction. However, the mechanism underlying microglial activation-induced neuroinflammation remains poorly understood. Ruxolitinib (RUX), a selective inhibitor of JAK1/2, was recently reported to inhibit inflammatory storms caused by SARS-CoV-2 in the lung. However, its role in disrupting inflammation post-SCI has not been confirmed. In this study, microglia were treated with RUX for 24 hours and then activated with interferon-γ for 6 hours. The results showed that interferon-γ-induced phosphorylation of JAK and STAT in microglia was inhibited, and the mRNA expression levels of pro-inflammatory cytokines tumor necrosis factor-α, interleukin-1β, interleukin-6, and cell proliferation marker Ki67 were reduced. In further in vivo experiments, a mouse model of spinal cord injury was treated intragastrically with RUX for 3 successive days, and the findings suggest that RUX can inhibit microglial proliferation by inhibiting the interferon-γ/JAK/STAT pathway. Moreover, microglia treated with RUX centripetally migrated toward injured foci, remaining limited and compacted within the glial scar, which resulted in axon preservation and less demyelination. Moreover, the protein expression levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6 were reduced. The neuromotor function of SCI mice also recovered. These findings suggest that RUX can inhibit neuroinflammation through inhibiting the interferon-γ/JAK/STAT pathway, thereby reducing secondary injury after SCI and producing neuroprotective effects.
Collapse
Affiliation(s)
- Zhan-Yang Qian
- Spine Center, Zhongda Hospital of Southeast University; Medical School, Southeast University, Nanjing, Jiangsu Province, China
| | - Ren-Yi Kong
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sheng Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Bin-Yu Wang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jie Chang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiang Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chao-Qin Wu
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zi-Yan Huang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ao Duan
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hai-Jun Li
- Department of Orthopedics, Hospital Affiliated 5 to Nantong University (Taizhou People's Hospital); Taizhou Clinical Medical School of Nanjing Medical University, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Lei Yang
- Department of Orthopedics, Hospital Affiliated 5 to Nantong University (Taizhou People's Hospital); Taizhou Clinical Medical School of Nanjing Medical University, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Xiao-Jian Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Wang Y, Pleasure D, Deng W, Guo F. Therapeutic Potentials of Poly (ADP-Ribose) Polymerase 1 (PARP1) Inhibition in Multiple Sclerosis and Animal Models: Concept Revisiting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102853. [PMID: 34935305 PMCID: PMC8844485 DOI: 10.1002/advs.202102853] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/12/2021] [Indexed: 05/05/2023]
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) plays a fundamental role in DNA repair and gene expression. Excessive PARP1 hyperactivation, however, has been associated with cell death. PARP1 and/or its activity are dysregulated in the immune and central nervous system of multiple sclerosis (MS) patients and animal models. Pharmacological PARP1 inhibition is shown to be protective against immune activation and disease severity in MS animal models while genetic PARP1 deficiency studies reported discrepant results. The inconsistency suggests that the function of PARP1 and PARP1-mediated PARylation may be complex and context-dependent. The article reviews PARP1 functions, discusses experimental findings and possible interpretations of PARP1 in inflammation, neuronal/axonal degeneration, and oligodendrogliopathy, three major pathological components cooperatively determining MS disease course and neurological progression, and points out future research directions. Cell type specific PARP1 manipulations are necessary for revisiting the role of PARP1 in the three pathological components prior to moving PARP1 inhibition into clinical trials for MS therapy.
Collapse
Affiliation(s)
- Yan Wang
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| | - David Pleasure
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityGuangzhou510006China
| | - Fuzheng Guo
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| |
Collapse
|
20
|
Song S, Guo R, Mehmood A, Zhang L, Yin B, Yuan C, Zhang H, Guo L, Li B. Liraglutide attenuate central nervous inflammation and demyelination through AMPK and pyroptosis-related NLRP3 pathway. CNS Neurosci Ther 2022; 28:422-434. [PMID: 34985189 PMCID: PMC8841291 DOI: 10.1111/cns.13791] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/22/2022] Open
Abstract
Aims Multiple sclerosis (MS) still maintains increasing prevalence and poor prognosis, while glucagon‐like peptide‐1 receptor (GLP‐1R) agonists show excellent neuroprotective capacities recently. Thus, we aim to evaluate whether the GLP‐1R agonist liraglutide (Lira) could ameliorate central nervous system demyelination and inflammation. Methods The therapeutic effect of Lira was tested on experimental autoimmune encephalitis (EAE) in vivo and a microglia cell line BV2 in vitro. Results Lira administration could ameliorate the disease score of EAE mice, delay the disease onset, ameliorate pathological demyelination and inflammation score in lumbar spinal cord, reduce pathogenic T helper cell transcription in spleen, restore phosphorylated adenosine monophosphate‐activated protein kinase (pAMPK) level, autophagy level, and inhibit pyroptosis‐related NLR family, pyrin domain‐containing protein 3 (NLRP3) pathway in lumbar spinal cord. Additionally, cell viability test, lactate dehydrogenase release test, and dead/live cell staining test for BV2 cells showed Lira could not salvage BV2 from nigericin‐induced pyroptosis significantly. Conclusion Lira has anti‐inflammation and anti‐demyelination effect on EAE mice, and the protective effect of Lira in the EAE model may be related to regulation of pAMPK pathway, autophagy, and NLRP3 pathway. However, Lira treatment cannot significantly inhibit pyroptosis of BV2 cells in vitro. Our study provides Lira as a potential candidate for Multiple Sclerosis treatment.
Collapse
Affiliation(s)
- Shuang Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Ruoyi Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Lu Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Bowen Yin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China.,Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Congcong Yuan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China.,Department of Neurology, Baoding First Central Hospital, Baoding, China
| | - Huining Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Li Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Bin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| |
Collapse
|
21
|
Protective effect and mechanism of nicotinamide adenine dinucleotide against optic neuritis in mice with experimental autoimmune encephalomyelitis. Int Immunopharmacol 2021; 98:107846. [PMID: 34174704 DOI: 10.1016/j.intimp.2021.107846] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 11/23/2022]
Abstract
Patients with multiple sclerosis (MS) are commonly accompanied by optic neuritis (ON) that causes retinal ganglion cell (RGC) death and even vision loss. Nicotinamide adenine dinucleotide (NAD+) can protect against cell apoptosis and attenuate MS-triggered symptoms. However, the effect of NAD+ on MS-triggered ON remains unclear. Herein, experimental autoimmune encephalomyelitis (EAE) was established by immunizing female C57BL/6 mice with MOG35-55 peptide. To investigate the effect of NAD+ on ON prevention and treatment, EAE mice received 250 mg/kg NAD+ daily via intraperitoneal injection after immunization and EAE onset, respectively. EX-527 (10 mg/kg, SIRT1 inhibitor) was intraperitoneally injected every two days to explore the role of SIRT1 in NAD+-induced therapeutic effect on EAE. NAD+ intervention attenuated the severity of EAE in mice. NAD+ intervention relieved inflammatory infiltration and CD3+ and CD4+ cell infiltration and decreased the number and activation of microglia and astrocytes in the optic nerve. NAD+ intervention also attenuated demyelination, axonal loss, oligodendrocyte apoptosis and oligodendrocyte progenitor cell recruitment and proliferation in the optic nerve and protected against RGC apoptosis in the retina. NAD+ intervention decreased pro-inflammatory cytokine mRNA and pro-apoptotic protein expression and enhanced anti-inflammatory cytokine mRNA expression and the SIRT1 signaling in the optic nerve and retina and regulated the Th1/Th17/Tregs immune response in the spleen. In addition, EX-527 reversed the therapeutic effect of NAD+ on EAE, suggesting that NAD+ prevented MS-triggered ON by activating the SIRT1 signaling pathway. This study shows the potential of NAD+ to be used as a drug in preventing and treating MS-related ON.
Collapse
|
22
|
Navarro MN, Gómez de Las Heras MM, Mittelbrunn M. Nicotinamide adenine dinucleotide metabolism in the immune response, autoimmunity and inflammageing. Br J Pharmacol 2021; 179:1839-1856. [PMID: 33817782 PMCID: PMC9292562 DOI: 10.1111/bph.15477] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolism is dynamically regulated to accompany immune cell function, and altered immunometabolism can result in impaired immune responses. Concomitantly, the pharmacological manipulation of metabolic processes offers an opportunity for therapeutic intervention in inflammatory disorders. The nicotinamide adenine dinucleotide (NAD+) is a critical metabolic intermediate that serves as enzyme cofactor in redox reactions, and is also used as a co‐substrate by many enzymes such as sirtuins, adenosine diphosphate ribose transferases and synthases. Through these activities, NAD+ metabolism regulates a broad spectrum of cellular functions such as energy metabolism, DNA repair, regulation of the epigenetic landscape and inflammation. Thus, the manipulation of NAD+ availability using pharmacological compounds such as NAD+ precursors can have immune‐modulatory properties in inflammation. Here, we discuss how the NAD+ metabolism contributes to the immune response and inflammatory conditions, with a special focus on multiple sclerosis, inflammatory bowel diseases and inflammageing.
Collapse
Affiliation(s)
- Maria N Navarro
- Interactions With The Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Manuel M Gómez de Las Heras
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Maria Mittelbrunn
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
23
|
Li Z, Liu Y, Jia A, Cui Y, Feng J. Cerebrospinal fluid cells immune landscape in multiple sclerosis. J Transl Med 2021; 19:125. [PMID: 33766068 PMCID: PMC7995713 DOI: 10.1186/s12967-021-02804-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Background Multiple Sclerosis (MS) is a potentially devastating autoimmune neurological disorder, which characteristically induces demyelination of white matter in the brain and spinal cord. Methods In this study, three characteristics of the central nervous system (CNS) immune microenvironment occurring during MS onset were explored; immune cell proportion alteration, differential gene expression profile, and related pathways. The raw data of two independent datasets were obtained from the ArrayExpress database; E-MTAB-69, which was used as a derivation cohort, and E-MTAB-2374 which was used as a validation cohort. Differentially expressed genes (DEGs) were identified by the false discovery rate (FDR) value of < 0.05 and |log2 (Fold Change)|> 1, for further analysis. Then, functional enrichment analyses were performed to explore the pathways associated with MS onset. The gene expression profiles were analyzed using CIBERSORT to identify the immune type alterations involved in MS disease. Results After verification, the proportion of five types of immune cells (plasma cells, monocytes, macrophage M2, neutrophils and eosinophils) in cerebrospinal fluid (CSF) were revealed to be significantly altered in MS cases compared to the control group. Thus, the complement and coagulation cascades and the systemic lupus erythematosus (SLE) pathways may play critical roles in MS. We identified NLRP3, LILRB2, C1QB, CD86, C1QA, CSF1R, IL1B and TLR2 as eight core genes correlated with MS. Conclusions Our study identified the change in the CNS immune microenvironment of MS cases by analysis of the in silico data using CIBERSORT. Our data may assist in providing directions for further research as to the molecular mechanisms of MS and provide future potential therapeutic targets in treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02804-7.
Collapse
Affiliation(s)
- Zijian Li
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yongchao Liu
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Aili Jia
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yueran Cui
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China.
| |
Collapse
|