1
|
Han X, Fu X, Guo W, Liu Y, Sun J, Wang T, Yang W. Ghrelin Inhibits Inflammasomes Activation in Astrocytes, Alleviates Pyroptosis, and Prevents Lipopolysaccharide-induced Depression-like Behavior in Mice. Inflammation 2024:10.1007/s10753-024-02190-4. [PMID: 39702621 DOI: 10.1007/s10753-024-02190-4] [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: 09/05/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
Depression is the leading cause of disability worldwide and places a significant burden on society. Neuroinflammation is closely associated with the pathophysiology of depression. Increasing evidence suggests that astrocytes, as the most abundant glial cells in the brain, are involved in the occurrence and development of depression due to morphological abnormalities and dysfunction. Astrocytes express the NOD-like receptor protein 2 (NLRP2) and NLRP3 inflammasomes, and the activation of inflammasomes induces pyroptosis. Ghrelin, a gastrointestinal peptide, plays vital role in regulating inflammation and alleviating stress. Therefore, we proposed a hypothesis that ghrelin inhibits the activation of inflammasomes on astrocytes, reduces pyroptosis, and consequently prevents depression. We used lipopolysaccharide (LPS)-induced mouse depression model and cultured primary astrocytes in vitro to explore the mechanism of the antidepressant effect of ghrelin. Our results showed that ghrelin effectively inhibited acute inflammatory responses and damage in the hippocampus and prefrontal cortex. The activation of NLRP2 and NLRP3 in astrocytes induced by LPS was significantly inhibited by ghrelin. Pretreatment with ghrelin effectively suppressed LPS-induced upregulation of pyroptosis-related proteins and mRNA. Ghrelin alleviated cell membrane pore formation and cell swelling, ultimately improved LPS-induced depression-like behavior. In vitro, ghrelin prevented the LPS-induced upregulation of pyroptosis-related proteins and mRNA expression in astrocytes, and inhibited the initiation and assembly of NLRP2 and NLRP3. Ghrelin exhibits antidepressant effects, inhibits inflammasomes activation in astrocytes, and prevents pyroptosis, suggesting a novel strategy for treating depression. This groundbreaking study reveals new avenues for targeting potential therapeutic interventions to alleviate depression.
Collapse
Affiliation(s)
- Xiaoou Han
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China.
| | - Xiying Fu
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Wanxu Guo
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Yaqi Liu
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Jiangjin Sun
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Tian Wang
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Wei Yang
- The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China.
| |
Collapse
|
2
|
Muhammad RN, Albahairy MA, Abd El Fattah MA, Ibrahim WW. Empagliflozin-activated AMPK elicits neuroprotective properties in reserpine-induced depression via regulating dynamics of hippocampal autophagy/inflammation and PKCζ-mediated neurogenesis. Psychopharmacology (Berl) 2024; 241:2565-2584. [PMID: 39158617 PMCID: PMC11569022 DOI: 10.1007/s00213-024-06663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024]
Abstract
RATIONALE Major depression has been an area of extensive research during the last decades, for it represents a leading cause of disability and suicide. The stark rise of depression rates influenced by life stressors, economic threats, pandemic era, and resistance to classical treatments, has made the disorder rather challenging. Adult hippocampal neurogenesis and plasticity are particularly sensitive to the dynamic interplay between autophagy and inflammation. In fact, the intricate balance between the two processes contributes to neuronal homeostasis and survival. OBJECTIVES Having demonstrated promising potentials in AMPK activation, a major metabolic sensor and autophagy regulator, empagliflozin (Empa) was investigated for possible antidepressant properties in the reserpine rat model of depression. RESULTS While the reserpine protocol elicited behavioral, biochemical, and histopathological changes relevant to depression, Empa outstandingly hindered these pathological perturbations. Importantly, hippocampal autophagic response markedly declined with reserpine which disrupted the AMPK/mTOR/Beclin1/LC3B machinery and, conversely, neuro-inflammation prevailed under the influence of the NLRP3 inflammasome together with oxidative/nitrative stress. Consequently, AMPK-mediated neurotrophins secretion obviously deteriorated through PKCζ/NF-κB/BDNF/CREB signal restriction. Empa restored hippocampal monoamines and autophagy/inflammation balance, driven by AMPK activation. By promoting the atypical PKCζ phosphorylation (Thr403) which subsequently phosphorylates NF-κB at Ser311, AMPK successfully reinforced BDNF/CREB signal and hippocampal neuroplasticity. The latter finding was supported by hippocampal CA3 toluidine blue staining to reveal intact neurons. CONCLUSION The current study highlights an interesting role for Empa as a regulator of autophagic and inflammatory responses in the pathology of depression. The study also pinpoints an unusual contribution for NF-κB in neurotrophins secretion via AMPK/PKCζ/NF-κB/BDNF/CREB signal transduction. Accordingly, Empa can have special benefits in diabetic patients with depressive symptoms. LIMITATIONS The influence of p-NF-κB (Ser311) on NLRP3 inflammasome assembly and activation has not been investigated, which can represent an interesting point for further research.
Collapse
Affiliation(s)
- Radwa N Muhammad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Mohammed A Albahairy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mai A Abd El Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
3
|
Filannino FM, Ruggiero M, Panaro MA, Lofrumento DD, Trotta T, Benameur T, Cianciulli A, Calvello R, Zoila F, Porro C. Irisin Attenuates Neuroinflammation Targeting the NLRP3 Inflammasome. Molecules 2024; 29:5623. [PMID: 39683782 DOI: 10.3390/molecules29235623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Neuroinflammation is defined as an immune response involving various cell types, particularly microglia, which monitor the neuroimmune axis. Microglia activate in two distinct ways: M1, which is pro-inflammatory and capable of inducing phagocytosis and releasing pro-inflammatory factors, and M2, which has anti-inflammatory properties. Inflammasomes are large protein complexes that form in response to internal danger signals, activating caspase-1 and leading to the release of pro-inflammatory cytokines such as interleukin 1β. Irisin, a peptide primarily released by muscles during exercise, was examined for its effects on BV2 microglial cells in vitro. Even at low concentrations, irisin was observed to influence the NLRP3 inflammasome, showing potential as a neuroprotective and anti-inflammatory agent after stimulation with lipopolysaccharides (LPSs). Irisin helped maintain microglia in their typical physiological state and reduced their migratory capacity. Irisin also increased Arg-1 protein expression, a marker of M2 polarization, while downregulating NLRP3, Pycard, caspase-1, IL-1β, and CD14. The results of this study indicate that irisin may serve as a crucial mediator of neuroprotection, thus representing an innovative tool for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Melania Ruggiero
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, I-73100 Lecce, Italy
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Environment, University of Bari, I-70125 Bari, Italy
| | - Federico Zoila
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, I-71100 Foggia, Italy
| |
Collapse
|
4
|
Li M, Ma L, Lv J, Zheng Z, Lu W, Yin X, Lin W, Wang P, Cui J, Hu L, Liu J. Design, synthesis, and biological evaluation of oridonin derivatives as novel NLRP3 inflammasome inhibitors for the treatment of acute lung injury. Eur J Med Chem 2024; 277:116760. [PMID: 39197252 DOI: 10.1016/j.ejmech.2024.116760] [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: 06/23/2024] [Revised: 07/31/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024]
Abstract
Acute lung injury (ALI) is a severe respiratory disorder closely associated with the excessive activation of the NLRP3 inflammasome. Oridonin (Ori), a natural diterpenoid compound, had been confirmed as a specific covalent NLRP3 inflammasome inhibitor, which was completely different from that of MCC950. However, the further clinical application of Ori was limited by its weak inhibitory activity against NLRP3 inflammasome (IC50 = 1240.67 nM). Fortunately, through systematic structure-optimization of Ori, D6 demonstrated the enhancement of IL-1β inhibitory activity (IC50 = 41.79 nM), which was better than the parent compound Ori. Then, by using SPR, molecular docking and MD simulation, D6 was verified to directly interact with NLRP3 via covalent and non-covalent interaction. The further anti-inflammatory mechanism studies were revealed that D6 could inhibit the activation of NLRP3 inflammasome without affecting the initiation phase of NLRP3 inflammasome activation, and D6 was a broad-spectrum and selective NLRP3 inflammasome inhibitor. Finally, D6 demonstrated a favorable therapeutic effect on LPS-induced ALI in mice model, and the potent pharmacodynamic effect of D6 was correlated with the specific inhibition of NLRP3 inflammasome activation in vivo. Thus, D6 is proved as a potent NLRP3 inhibitor, and has the potential to develop as a novel anti-ALI agent.
Collapse
Affiliation(s)
- Mengting Li
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingyu Ma
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiahao Lv
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhe Zheng
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenyu Lu
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xunkai Yin
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weijiang Lin
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ping Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian Cui
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jian Liu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
5
|
Wei B, Li H, Cheng M, Yang Y, Liu B, Tian Y, Sun Y, Liu T, She R, Tian J. NLRP3 Inflammasome Activation Mediates Hepatitis E Virus-Induced Neuroinflammation. J Viral Hepat 2024; 31:729-738. [PMID: 39136210 DOI: 10.1111/jvh.13998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/17/2024] [Indexed: 10/23/2024]
Abstract
Hepatitis E virus (HEV) is a foodborne zoonotic pathogen that is supposed to be one of the most common causes of acute viral hepatitis. However, HEV infection has been recently associated with a wide spectrum of extrahepatic manifestations, particularly neurological disorders. Previous studies have shown that HEV is able to cross the blood-brain barrier (BBB) and induce inflammatory response of the central nervous system. However, the pathogenesis of HEV-induced neuroinflammation and tissue injury of the central nervous system have yet to be fully elucidated. In this study, activation of NLRP3 inflammasome following HEV infection were investigated. In a gerbil model infected by HEV, brain histopathological changes including gliosis, neuronophagia and neuron injury were observed and expression of NLRP3, caspase-1, IL-1β and IL-18 were elevated. Brain microvascular endothelial cells (BMECs) are key components of the BBB that protects the brain from various challenges. Following HEV infection, virus-like particles range from 30 to 40 nm in diameter were observed in human BMECs (hBMECs). Enhanced expression levels of NLRP3 and subsequent ASC, caspase-1, IL-1β and IL-18 were detected in infected cells. Treatment with MCC950 alleviated HEV infection induced activation of NLRP3 inflammasome, mitochondrial damage and VE-cadherin degradation. The findings provide new insights into HEV-associated neuroinflammation. Moreover, targeting NLRP3 inflammasome signalling is a promising therapeutic in HEV-induced neurological disorder.
Collapse
Affiliation(s)
- Bingyan Wei
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Huopeng Li
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Minheng Cheng
- Beijing Center for Animal Disease Control and Prevention, Beijing, China
| | - Yifei Yang
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Bo Liu
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuewei Tian
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yaxin Sun
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tianlong Liu
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ruiping She
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jijing Tian
- Laboratory of Animal Pathology and Public Health, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
6
|
Park S, Cho S, Kim KM, Chu MK, Kim CH, Jeong KH, Kim WJ. Honokiol-induced SIRT3 upregulation protects hippocampal neurons by suppressing inflammatory processes in pilocarpine-induced status epilepticus. Neurochem Int 2024; 180:105873. [PMID: 39362498 DOI: 10.1016/j.neuint.2024.105873] [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: 05/27/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Status epilepticus (SE), a continuous and self-sustaining epileptic seizure lasting more than 30 min, is a neurological emergency that can cause severe brain injuries and increase the risk for the development of epilepsy. Over the past few decades, accumulating evidence has suggested the importance of brain inflammation in the pathogenesis of epilepsy. Honokiol (HNK), a pharmacological activator of sirtuin 3 (SIRT3), is a bioactive compound extracted from the bark or leaves of Magnolia plants that possesses therapeutic benefits for preventing the development of inflammatory injury. However, the therapeutic effects of HNK against epileptic brain injury via regulating molecular mechanisms related to neuroinflammation remains elusive. Therefore, the present study investigated the effects of HNK on pilocarpine-induced status epilepticus (PCSE) and the therapeutic benefits of HNK in regulating inflammatory processes in the hippocampus. Treatment with HNK before PCSE induction attenuated the initiation of behavioral seizures. Post-treatment with HNK after SE onset increased SIRT3 expression, which mitigated glial activation, including reactive astrocytes and activated microglia, in the hippocampus following PCSE. Moreover, HNK treatment reduced the activation of the nuclear factor-κB/nucleotide-binding domain leucine-rich repeat with a pyrin-domain containing 3 inflammasome pathway, thereby inhibiting the production of interleukin-1β pro-inflammatory cytokine, subsequently alleviating PCSE-triggered apoptotic neuronal death in the hippocampus. These results indicate that HNK-induced SIRT3 upregulation has the potential to prevent the progression of epileptic neuropathology through its anti-inflammatory properties. Therefore, the present study suggests that HNK is a natural therapeutic agent for epileptic brain injury.
Collapse
Affiliation(s)
- Soojin Park
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Soomi Cho
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Kyung Min Kim
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Min Kyung Chu
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chul Hoon Kim
- Department of Pharmacology, Brain Korea 21 Project, Brain Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Kyoung Hoon Jeong
- Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Won-Joo Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea.
| |
Collapse
|
7
|
Kulkarni R, Kumari S, Dhapola R, Sharma P, Singh SK, Medhi B, HariKrishnaReddy D. Association Between the Gut Microbiota and Alzheimer's Disease: An Update on Signaling Pathways and Translational Therapeutics. Mol Neurobiol 2024:10.1007/s12035-024-04545-2. [PMID: 39460901 DOI: 10.1007/s12035-024-04545-2] [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: 05/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Alzheimer's disease (AD) is a cognitive disease with high morbidity and mortality. In AD patients, the diversity of the gut microbiota is altered, which influences pathology through the gut-brain axis. Probiotic therapy alleviates pathological and psychological consequences by restoring the diversity of the gut microbial flora. This study addresses the role of altered gut microbiota in the progression of neuroinflammation, which is a major hallmark of AD. This process begins with the activation of glial cells, leading to the release of proinflammatory cytokines and the modulation of cholinergic anti-inflammatory pathways. Short-chain fatty acids, which are bacterial metabolites, provide neuroprotective effects and maintain blood‒brain barrier integrity. Furthermore, the gut microbiota stimulates oxidative stress and mitochondrial dysfunction, which promote AD progression. The signaling pathways involved in gut dysbiosis-mediated neuroinflammation-mediated promotion of AD include cGAS-STING, C/EBPβ/AEP, RAGE, TLR4 Myd88, and the NLRP3 inflammasome. Preclinical studies have shown that natural extracts such as Ganmaidazao extract, isoorentin, camelia oil, Sparassis crispa-1, and xanthocerasides improve gut health and can delay the worsening of AD. Clinical studies using probiotics such as Bifidobacterium spp., yeast beta-glucan, and drugs such as sodium oligomannate and rifaximine have shown improvements in gut health, resulting in the amelioration of AD symptoms. This study incorporates the most current research on the pathophysiology of AD involving the gut microbiota and highlights the knowledge gaps that need to be filled to develop potent therapeutics against AD.
Collapse
Affiliation(s)
- Rutweek Kulkarni
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sunil K Singh
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India.
| |
Collapse
|
8
|
Salim Abed H, Oghenemaro EF, Kubaev A, Jeddoa ZMA, S R, Sharma S, Vashishth R, Jabir MS, Jawad SF, Zwamel AH. Non-coding RNAs as a Critical Player in the Regulation of Inflammasome in Inflammatory Bowel Diseases; Emphasize on lncRNAs. Cell Biochem Biophys 2024:10.1007/s12013-024-01585-2. [PMID: 39424765 DOI: 10.1007/s12013-024-01585-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2024] [Indexed: 10/21/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic disease caused by a dysregulated immune response to host intestinal microflora. A hyperactive inflammatory and immunological response in the gut has been shown to be one of the disease's long-term causes despite the complexity of the clinical pathology of IBD. The innate immune system activator known as human gut inflammasome is thought to be a significant underlying cause of pathology and is closely linked to the development of IBD. It is essential to comprehend the function of inflammasome activation in IBD to treat it effectively. Systemic inflammasome regulation may be a proper therapeutic and clinical strategy to manage IBD symptoms since inflammasomes may have a significant function in IBD. Non-coding RNAs (ncRNAs) are a type of RNA transcript that is incapable of encoding proteins or peptides. In IBD, inflammation develops and worsens as a result of its imbalance. Culminating evidence has been shown that ncRNAs, and particularly long non-coding RNAs (lncRNAs), may play a role in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in IBD. The relationship between IBD and the gut inflammasome, as well as current developments in IBD research and treatment approaches, have been the main topics of this review. We have covered inflammasomes and their constituents, results from in vivo research, inflammasome inhibitors, and advancements in inflammasome-targeted therapeutics for IBD.
Collapse
Affiliation(s)
- Hussein Salim Abed
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Ramadi, Iraq
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria.
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, 140100, Uzbekistan
| | | | - RenukaJyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Karbala, Iraq
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001, Babil, Iraq
| | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
9
|
Wang H, Wang X, Wang H, Shao S, Zhu J. Chronic Corticosterone Administration-Induced Mood Disorders in Laboratory Rodents: Features, Mechanisms, and Research Perspectives. Int J Mol Sci 2024; 25:11245. [PMID: 39457027 PMCID: PMC11508944 DOI: 10.3390/ijms252011245] [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: 09/09/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Mood disorders mainly affect the patient's daily life, lead to suffering and disability, increase the incidence rate of many medical illnesses, and even cause a trend of suicide. The glucocorticoid (GC)-mediated hypothalamus-pituitary-adrenal (HPA) negative feedback regulation plays a key role in neuropsychiatric disorders. The balance of the mineralocorticoid receptor (MR)/glucocorticoid receptor (GR) level contributes to maintaining the homeostasis of the neuroendocrine system. Consistently, a chronic excess of GC can also lead to HPA axis dysfunction, triggering anxiety, depression, memory loss, and cognitive impairment. The animal model induced by chronic corticosterone (CORT) administration has been widely adopted because of its simple replication and strong stability. This review summarizes the behavioral changes and underlying mechanisms of chronic CORT administration-induced animal models, including neuroinflammatory response, pyroptosis, oxidative stress, neuroplasticity, and apoptosis. Notably, CORT administration at different doses and cycles can destroy the balance of the MR/GR ratio to make dose-dependent effects of CORT on the central nervous system (CNS). This work aims to offer an overview of the topic and recommendations for future cognitive function research.
Collapse
Affiliation(s)
- Hao Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (H.W.); (X.W.); (H.W.); (S.S.)
| | - Xingxing Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (H.W.); (X.W.); (H.W.); (S.S.)
| | - Huan Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (H.W.); (X.W.); (H.W.); (S.S.)
| | - Shuijin Shao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (H.W.); (X.W.); (H.W.); (S.S.)
| | - Jing Zhu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (H.W.); (X.W.); (H.W.); (S.S.)
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai 201108, China
| |
Collapse
|
10
|
Cai W, Wei XF, Zhang JR, Tao L, Li D, Zhang K, Shen WD. Acupuncture ameliorates depression-like behavior of poststroke depression model rats through the regulation of gut microbiota and NLRP3 inflammasome in the colon. Neuroreport 2024; 35:883-894. [PMID: 39207304 DOI: 10.1097/wnr.0000000000002076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
This study was conducted to examine the effects of acupuncture on gut microbiota and expression of NLRP3 inflammasome in the colon in poststroke depression (PSD) model rats. Sprague-Dawley male rats were randomized into four groups: sham surgery group, poststroke depression group, acupuncture group, and probiotics group. Acupuncture therapy at Baihui (GV20), Shenting (GV24), bilateral Zusanli (ST36) acupoints in the acupuncture group and probiotic gavage therapy in the probiotics group were performed once per day for 2 weeks. Behaviors of depression were assessed by using weight measurements, sucrose preference test, open field test, and forced swimming test. Histopathological alterations in the colon were determined by hematoxylin-eosin staining, the expression of NLRP3/ASC/caspase-1 pathway-related proteins was analyzed by western blotting. Serum levels of IL-1β and IL-18 were derived from ELISA. The 16S rRNA gene sequencing was performed to examine and analyze the differences of gut microbiota of rats among all groups. Acupuncture was effective to increase weight and ameliorate depressive-like behaviors in PSD rats. Acupuncture increased the diversity of gut microbiota, upregulated the abundance of Bifidobacteriaceae and Lactobacillaceae, and decreased the relative abundance of Peptostreptococcaceae, Rikenellaceae, Eggerthellaceae, and Streptococcaceae at family level. Acupuncture effectively improved the pathological changes in the colon. Meanwhile, acupuncture reduced NLRP3, ASC, caspase-1 protein expressions in the colon, and serum levels of IL-18 and IL-1β. Acupuncture may reduce depressive-like behaviors of PSD by regulating the gut microbiota and suppressing hyperactivation of NLRP3 inflammasome in the colon. Microbiota-gut-brain axis may be an effective target pathway for acupuncture treatment of PSD.
Collapse
Affiliation(s)
- Wa Cai
- Department of Acupuncture, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai
| | - Xi-Fang Wei
- Department of Acupuncture, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou
| | - Jing-Ruo Zhang
- Department of Acupuncture and Moxibustion, Jiaxing Hospital of TCM, Zhejiang Chinese Medicine University, Jiaxing, China
| | - Larissa Tao
- Department of Acupuncture, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai
| | - Dong Li
- Department of Acupuncture, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai
| | - Kun Zhang
- Department of Acupuncture, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai
| | - Wei-Dong Shen
- Department of Acupuncture, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai
| |
Collapse
|
11
|
Taylor RR, Keane RW, Guardiola B, López-Lage S, Moratinos L, Dietrich WD, Perez-Barcena J, de Rivero Vaccari JP. Inflammasome Proteins Are Reliable Biomarkers of the Inflammatory Response in Aneurysmal Subarachnoid Hemorrhage. Cells 2024; 13:1370. [PMID: 39195261 DOI: 10.3390/cells13161370] [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: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is caused by abnormal blood vessel dilation and subsequent rupture, resulting in blood pooling in the subarachnoid space. This neurological insult results in the activation of the inflammasome, a multiprotein complex that processes pro-inflammatory interleukin (IL)-1 cytokines leading to morbidity and mortality. Moreover, increases in inflammasome proteins are associated with clinical deterioration in many neurological diseases. Limited studies have investigated inflammasome protein expression following aSAH. Reliable markers of the inflammatory response associated with aSAH may allow for earlier detection of patients at risk for complications and aid in the identification of novel pharmacologic targets. Here, we investigated whether inflammasome signaling proteins may serve as potential biomarkers of the inflammatory response in aSAH. Serum and cerebrospinal fluid (CSF) from fifteen aSAH subjects and healthy age-matched controls and hydrocephalus (CSF) no-aneurysm controls were evaluated for levels of inflammasome signaling proteins and downstream pro-inflammatory cytokines. Protein measurements were carried out using Simple Plex and Single-Molecule Array (Simoa) technology. The area under the curve (AUC) was calculated using receiver operating characteristics (ROCs) to obtain information on biomarker reliability, specificity, sensitivity, cut-off points, and likelihood ratio. In addition, a Spearman r correlation matrix was performed to determine the correlation between inflammasome protein levels and clinical outcome measures. aSAH subjects demonstrated elevated caspase-1, apoptosis-associated speck-like protein with a caspase recruiting domain (ASC), IL-18 and IL-1β levels in serum, and CSF when compared to controls. Each of these proteins was found to be a promising biomarker of inflammation in aSAH in the CSF. In addition, ASC, caspase-1, and IL-1β were found to be promising biomarkers of inflammation in aSAH in serum. Furthermore, we found that elevated levels of inflammasome proteins in serum are useful to predict worse functional outcomes following aSAH. Thus, the determination of inflammasome protein levels in CSF and serum in aSAH may be utilized as reliable biomarkers of inflammation in aSAH and used clinically to monitor patient outcomes.
Collapse
Affiliation(s)
- Ruby R Taylor
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Medical Scientist Training Program, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Robert W Keane
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Cellular Physiology and Molecular Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Begoña Guardiola
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain
| | - Sofía López-Lage
- Neurosurgical Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain
| | - Lesmes Moratinos
- Neurosurgical Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jon Perez-Barcena
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Cellular Physiology and Molecular Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
12
|
Jin GN, Wang Y, Liu YM, Lu YN, Lu JM, Wang JH, Ma JW, Quan YZ, Gao HY, Cui YX, Xu X, Piao LX. Arctiin Mitigates Neuronal Injury by Modulating the P2X7R/NLPR3 Inflammasome Signaling Pathway. Inflammation 2024:10.1007/s10753-024-02117-z. [PMID: 39154088 DOI: 10.1007/s10753-024-02117-z] [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: 05/21/2024] [Revised: 06/13/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
Depression, recognized globally as a primary cause of disability, has its pathogenesis closely related to neuroinflammation and neuronal damage. Arctiin (ARC), the major bioactive component of Fructus arctii, has various pharmacological activities, such as anti-inflammatory and neuroprotective effects. Building on previous findings that highlighted ARC's capability to mitigate depression by dampening microglial hyperactivation and thereby reducing neuroinflammatory responses and cortical neuronal damage in mice, the current study delves deeper into ARC's therapeutic potential by examining its impact on hippocampal neuronal damage in depression. Utilizing both chronic unpredictable mild stress (CUMS)-induced depression model in mice and corticosterone (CORT)-stimulated PC12 cell model of neuronal damage, the techniques including Nissl staining, immunohistochemistry, western blotting, ELISA, lactate dehydrogenase assays, colony formation assays, immunofluorescence staining and molecular docking were employed to unravel the mechanisms behind ARC's neuroprotective effects. The findings revealed that ARC not only mitigates hippocampal neuropathological damage and reduces serum CORT levels in CUMS-exposed mice but also enhances cell activity while reducing lactate dehydrogenase release in CORT-stimulated PC12 cells. ARC attenuated neuroinflammatory responses and neuronal apoptosis by inhibiting the overactivation of the P2X7 receptor (P2X7R)/NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathway, similar to the effect of A438079 (P2X7R antagonist). Interestingly, pretreatment with A438079 blocked the neuroprotective effect of ARC. Computer modeling predicted that both ARC and A438079 have strong binding with P2X7R and they have the same binding site. These results suggested that ARC may exert a neuroprotective role by binding to P2X7R, thereby inhibiting the P2X7R/NLRP3 inflammasome signaling pathway.
Collapse
Affiliation(s)
- Guang-Nan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yu Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yi-Ming Liu
- Department of Neurology, Yanbian University Hospital, Yanbian University, Yanji, 133000, Jilin Province, China
| | - Yu-Nan Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-Mei Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-He Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jing-Wen Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yan-Zhu Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Hong-Yan Gao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Yue-Xian Cui
- Department of Neurology, Yanbian University Hospital, Yanbian University, Yanji, 133000, Jilin Province, China.
| | - Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
13
|
Chen Z, Shang Y, Ou Y, Shen C, Cao Y, Hu H, Yang R, Liu T, Liu Q, Song M, Zong D, Xiang X, Peng Y, Ouyang R. Obstructive Sleep Apnea Plasma-Derived Exosomes Mediate Cognitive Impairment Through Hippocampal Neuronal Cell Pyroptosis. Am J Geriatr Psychiatry 2024; 32:922-939. [PMID: 38290937 DOI: 10.1016/j.jagp.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/25/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVE Obstructive sleep apnea (OSA) is associated with impaired cognitive function. Exosomes are secreted by most cells and play a role in OSA-associated cognitive impairment (CI). The aim of this study was to investigate whether OSA plasma-derived exosomes cause CI through hippocampal neuronal cell pyroptosis, and to identify exosomal miRNAs in OSA plasma-derived. MATERIALS AND METHODS Plasma-derived exosomes were isolated from patients with severe OSA and healthy comparisons. Daytime sleepiness and cognitive function were assessed using the Epworth Sleepiness Scale (ESS) and the Beijing version of the Montreal Cognitive Assessment Scale (MoCA). Exosomes were coincubated with mouse hippocampal neurons (HT22) cells to evaluate the effect of exosomes on pyroptosis and inflammation of HT22 cells. Meanwhile, exosomes were injected into C57BL/6 male mice via caudal vein, and then morris water maze was used to evaluate the spatial learning and memory ability of the mice, so as to observe the effects of exosomes on the cognitive function of the mice. Western blot and qRT-PCR were used to detect the expressions of Gasdermin D (GSDMD) and Caspase-1 to evaluate the pyroptosis level. The expression of IL-1β, IL-6, IL-18 and TNF-α was detected by qRT-PCR to assess the level of inflammation. Correlations of GSDMD and Caspase-1 expression with clinical parameters were evaluated using Spearman's rank correlation analysis. In addition, plasma exosome miRNAs profile was identified, followed by Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. RESULTS Compared to healthy comparisons, body mass index (BMI), apnea-hypopnea index (AHI), oxygen desaturation index (ODI), and ESS scores were increased in patients with severe OSA, while lowest oxygen saturation during sleep (LSaO2), mean oxygen saturation during sleep (MSaO2) and MoCA scores were decreased. Compared to the PBS group (NC) and the healthy comparison plasma-derived exosomes (NC-EXOS), the levels of GSDMD and Caspase-1 and IL-1β, IL-6, IL-18 and TNF-α were increased significantly in the severe OSA plasma-derived exosomes (OSA-EXOS) coincubated with HT22 cells. Compared to the NC and NC-EXOS groups, the learning and memory ability of mice injected with OSA-EXOS was decreased, and the expression of GSDMD and Caspase-1 in hippocampus were significantly increased, along with the levels of IL-1β, IL-6, IL-18 and TNF-α. Spearman correlation analysis found that clinical AHI in HCs and severe OSA patients was positively correlated with GSDMD and Caspase-1 in HT22 cells from NC-EXOS and OSA-EXOS groups, while negatively correlated with clinical MoCA. At the same time, clinical MoCA in HCs and severe OSA patients was negatively correlated with GSDMD and Caspase-1 in HT22 cells from NC-EXOS and OSA-EXOS groups. A unique exosomal miRNAs profile was identified in OSA-EXOS group compared to the NC-EXOS group, in which 28 miRNAs were regulated and several KEGG and GO pathways were identified. CONCLUSIONS The results of this study show a hypothesis that plasma-derived exosomes from severe OSA patients promote pyroptosis and increased expression of inflammatory factors in vivo and in vitro, and lead to impaired cognitive function in mice, suggesting that OSA-EXOS can mediate CI through pyroptosis of hippocampal neurons. In addition, exosome cargo from OSA-EXOS showed a unique miRNAs profile compared to NC-EXOS, suggesting that plasma exosome associated miRNAs may reflect the differential profile of OSA related diseases, such as CI.
Collapse
Affiliation(s)
- Zhifeng Chen
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Yulin Shang
- Ophthalmology and Otorhinolaryngology (YS), Zigui County Traditional Chinese Medicine Hospital, Zigui, China
| | - Yanru Ou
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Chong Shen
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Ying Cao
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Hui Hu
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Ruibing Yang
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Ting Liu
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Qingqing Liu
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Min Song
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Dandan Zong
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China
| | - Xudong Xiang
- Department of Emergency (XX), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yating Peng
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China.
| | - Ruoyun Ouyang
- Department of Respiratory Medicine (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Unit of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Pulmonary (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Critical Care Medicine in Hunan Province, Changsha, Hunan, China; Diagnosis and Treatment Center of Respiratory Disease (ZC, YO, CS, YC, HH, RY, TL, QL, MS, DZ, YP, RO), Central South University, Changsha, Hunan, China.
| |
Collapse
|
14
|
Luo E, Li Z, Zhang S, Wen Y, Yang Z, Zeng H, Ding H. Hyperglycemia induces microglial pyroptosis by increasing oxygen extraction rate: Implication in neurological impairment during ischemic stroke. Mol Med Rep 2024; 30:146. [PMID: 38940333 PMCID: PMC11222914 DOI: 10.3892/mmr.2024.13270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Elevated levels of blood glucose in patients with ischemic stroke are associated with a worse prognosis. The present study aimed to explore whether hyperglycemia promotes microglial pyroptosis by increasing the oxygen extraction rate in an acute ischemic stroke model. C57BL/6 mice that underwent middle cerebral artery occlusion were used for assessment of blood glucose level and neurological function. The cerebral oxygen extraction ratio (CERO2), oxygen consumption rate (OCR) and partial pressure of brain tissue oxygen (PbtO2) were measured. To investigate the significance of the NOD‑like receptor protein 3 (NLRP3) inflammasome, NLRP3‑/‑ mice were used, and the expression levels of NLRP3, caspase‑1, full‑length gasdermin D (GSDMD‑FL), GSDMD‑N domain (GSDMD‑N), IL‑1β and IL‑18 were evaluated. In addition, Z‑YVAD‑FMK, a caspase‑1 inhibitor, was used to treat microglia to determine whether activation of the NLRP3 inflammasome was required for the enhancing effect of hyperglycemia on pyroptosis. It was revealed that hyperglycemia accelerated cerebral injury in the acute ischemic stroke model, as evidenced by decreased latency to fall and the percentage of foot fault. Hyperglycemia aggravated hypoxia by increasing the oxygen extraction rate, as evidenced by increased CERO2 and OCR, and decreased PbtO2 in response to high glucose treatment. Furthermore, hyperglycemia‑induced microglial pyroptosis was confirmed by detection of increased levels of caspase‑1, GSDMD‑N, IL‑1β and IL‑18 and a decreased level of GSDMD‑FL. However, the knockout of NLRP3 attenuated these effects. Pharmacological inhibition of caspase‑1 also reduced the expression levels of GSDMD‑N, IL‑1β and IL‑18 in microglial cells. These results suggested that hyperglycemia stimulated NLRP3 inflammasome activation by increasing the oxygen extraction rate, thus leading to the aggravation of pyroptosis following ischemic stroke.
Collapse
Affiliation(s)
- Ensi Luo
- Department of Endocrinology, Binhaiwan Central Hospital of Dongguan, Dongguan Hospital Affiliated to Medical College of Jinan University, Dongguan, Guangdong 523903, P.R. China
| | - Zhuo Li
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Shiying Zhang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Yin Wen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Zixi Yang
- College of Continuing Education, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Hongke Zeng
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
15
|
Diz-Chaves Y, Maastor Z, Spuch C, Lamas JA, González-Matías LC, Mallo F. Glucagon-like peptide 1 receptor activation: anti-inflammatory effects in the brain. Neural Regen Res 2024; 19:1671-1677. [PMID: 38103230 PMCID: PMC10960307 DOI: 10.4103/1673-5374.389626] [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: 01/16/2023] [Revised: 09/08/2023] [Accepted: 10/14/2023] [Indexed: 12/18/2023] Open
Abstract
The glucagon-like peptide 1 is a pleiotropic hormone that has potent insulinotropic effects and is key in treating metabolic diseases such as diabetes and obesity. Glucagon-like peptide 1 exerts its effects by activating a membrane receptor identified in many tissues, including different brain regions. Glucagon-like peptide 1 activates several signaling pathways related to neuroprotection, like the support of cell growth/survival, enhancement promotion of synapse formation, autophagy, and inhibition of the secretion of proinflammatory cytokines, microglial activation, and apoptosis during neural morphogenesis. The glial cells, including astrocytes and microglia, maintain metabolic homeostasis and defense against pathogens in the central nervous system. After brain insult, microglia are the first cells to respond, followed by reactive astrocytosis. These activated cells produce proinflammatory mediators like cytokines or chemokines to react to the insult. Furthermore, under these circumstances, microglia can become chronically inflammatory by losing their homeostatic molecular signature and, consequently, their functions during many diseases. Several processes promote the development of neurological disorders and influence their pathological evolution: like the formation of protein aggregates, the accumulation of abnormally modified cellular constituents, the formation and release by injured neurons or synapses of molecules that can dampen neural function, and, of critical importance, the dysregulation of inflammatory control mechanisms. The glucagon-like peptide 1 receptor agonist emerges as a critical tool in treating brain-related inflammatory pathologies, restoring brain cell homeostasis under inflammatory conditions, modulating microglia activity, and decreasing the inflammatory response. This review summarizes recent advances linked to the anti-inflammatory properties of glucagon-like peptide 1 receptor activation in the brain related to multiple sclerosis, Alzheimer's disease, Parkinson's disease, vascular dementia, or chronic migraine.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- Biomedical Research Centre (CINBIO), Laboratory of Endocrinology, University of Vigo, Galicia Sur Health Research Institute, Vigo, Spain
| | - Zainab Maastor
- Biomedical Research Centre (CINBIO), Laboratory of Endocrinology, University of Vigo, Galicia Sur Health Research Institute, Vigo, Spain
| | - Carlos Spuch
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, CIBERSAM, Hospital Álvaro Cunqueiro, Sala Investigación, Estrada Clara Campoamor, Vigo, Spain
| | - José Antonio Lamas
- Biomedical Research Centre (CINBIO), Laboratory of Neuroscience, University of Vigo, Galicia Sur Health Research Institute, Vigo, Spain
| | - Lucas C. González-Matías
- Biomedical Research Centre (CINBIO), Laboratory of Endocrinology, University of Vigo, Galicia Sur Health Research Institute, Vigo, Spain
| | - Federico Mallo
- Biomedical Research Centre (CINBIO), Laboratory of Endocrinology, University of Vigo, Galicia Sur Health Research Institute, Vigo, Spain
| |
Collapse
|
16
|
Brint A, Greene S, Fennig-Victor AR, Wang S. Multiple sclerosis: the NLRP3 inflammasome, gasdermin D, and therapeutics. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:62. [PMID: 39118955 PMCID: PMC11304424 DOI: 10.21037/atm-23-1960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/26/2024] [Indexed: 08/10/2024]
Abstract
Multiple sclerosis (MS) stands as a chronic inflammatory disease characterized by its neurodegenerative impacts on the central nervous system. The complexity of MS and the significant challenges it poses to patients have made the exploration of effective treatments a crucial area of research. Among the various mechanisms under investigation, the role of inflammation in MS progression is of particular interest. Inflammatory responses within the body are regulated by various cellular mechanisms, one of which involves the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat (LRR)-, and pyrin domains (PYD)-containing protein 3 (NLRP3). NLRP3 acts as a sensor within cells, playing a pivotal role in controlling the inflammatory response. Its activation is a critical step leading to the assembly of the NLRP3 inflammasome complex, a process that has profound implications for inflammatory diseases like MS. The NLRP3 inflammasome's activation is intricately linked to the subsequent activation of caspase 1 and gasdermin D (GsdmD), signaling pathways that are central to the inflammatory process. GsdmD, a prominent member of the Gasdermin protein family, is particularly noteworthy for its role in pyroptotic cell death, a form of programmed cell death that is distinct from apoptosis and is characterized by its inflammatory nature. This pathway's activation contributes significantly to the pathology of MS by exacerbating inflammatory responses within the nervous system. Given the detrimental effects of unregulated inflammation in MS, therapeutics targeting these inflammatory processes offer a promising avenue for alleviating the symptoms experienced by patients. This review delves into the intricacies of the pyroptotic pathways, highlighting how the formation of the NLRP3 inflammasome induces such pathways and the potential intervention points for therapeutic agents. By inhibiting key steps within these pathways, it is possible to mitigate the inflammatory response, thereby offering relief to those suffering from MS. Understanding these mechanisms not only sheds light on the pathophysiology of MS but also paves the way for the development of novel therapeutic strategies aimed at controlling the disease's progression through the modulation of the body's inflammatory response.
Collapse
Affiliation(s)
- Amie Brint
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, USA
- College of Medicine and Graduate School, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Seth Greene
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| | - Alyssa R. Fennig-Victor
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| | - Shanzhi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| |
Collapse
|
17
|
Wang Q, Xie Y, Ma S, Luo H, Qiu Y. Role of microglia in diabetic neuropathic pain. Front Cell Dev Biol 2024; 12:1421191. [PMID: 39135776 PMCID: PMC11317412 DOI: 10.3389/fcell.2024.1421191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Approximately one-third of the patients with diabetes worldwide suffer from neuropathic pain, mainly categorized by spontaneous and stimulus-induced pain. Microglia are a class of immune effector cells residing in the central nervous system and play a pivotal role in diabetic neuropathic pain (DNP). Microglia specifically respond to hyperglycemia along with inflammatory cytokines and adenosine triphosphate produced during hyperglycemic damage to nerve fibers. Because of the presence of multiple receptors on the microglial surface, microglia are dynamically and highly responsive to their immediate environment. Following peripheral sensitization caused by hyperglycemia, microglia are affected by the cascade of inflammatory factors and other substances and respond accordingly, resulting in a change in their functional state for DNP pathogenesis. Inhibition of receptors such as P2X reporters, reducing cytokine expression levels in the microglial reactivity mechanisms, and inhibiting their intracellular signaling pathways can effectively alleviate DNP. A variety of drugs attenuate DNP by inhibiting the aforementioned processes induced by microglial reactivity. In this review, we summarize the pathological mechanisms by which microglia promote and maintain DNP, the drugs and therapeutic techniques available, and the latest advances in this field.
Collapse
Affiliation(s)
- Qian Wang
- Department of Endocrinology and Metabolism, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yilin Xie
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shichao Ma
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yue Qiu
- Department of Endocrinology and Metabolism, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| |
Collapse
|
18
|
Zhang L, Li X, Gao H, Chang W, Li P. Gut microbiota-lncRNA/circRNA crosstalk: implications for different diseases. Crit Rev Microbiol 2024:1-15. [PMID: 38967384 DOI: 10.1080/1040841x.2024.2375516] [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: 07/04/2023] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
The gut microbiota features an abundance of diverse microorganisms and represents an important component of human physiology and metabolic homeostasis, indicating their roles in a wide array of physiological and pathological processes in the host. Maintaining balance in the gut microbiota is critical for normal functionality as microbial dysbiosis can lead to the occurrence and development of diseases through various mechanisms. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are non-coding RNAs that perform important regulatory functions for many processes. Furthermore, the gut microbiota and lncRNAs/circRNAs are known to interact in a range of both physiological and pathological activities. In this article, we review existing research relevant to the interaction between the gut microbiota and lncRNAs/circRNAs and investigate the role of their crosstalk in the pathogenesis of different diseases. Studies have shown that, the gut microbiota can target lncRNAs ENO1-IT1, BFAL1, and LINC00152 to regulate colorectal cancer development via various signaling pathways. In addition, the gut microbiota can influence mental diseases and lung tumor metastasis by modulating circRNAs such as circNF1-419, circ_0001239, circHIPK2 and mmu_circ_0000730. These findings provide a theoretical basis for disease prevention and treatment and suggest that gut microbiota-lncRNA/circRNA crosstalk has high clinical value.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Huijuan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Shen J, Xu J, Wen Y, Tang Z, Li J, Sun J. Carnosine ameliorates postoperative cognitive dysfunction of aged rats by limiting astrocytes pyroptosis. Neurotherapeutics 2024; 21:e00359. [PMID: 38664193 PMCID: PMC11301240 DOI: 10.1016/j.neurot.2024.e00359] [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: 11/08/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 07/15/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common postoperative complication in elderly patients, and neuroinflammation is a key hallmark. Recent studies suggest that the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-mediated astrocytes pyroptosis is involved in the regulation of neuroinflammation in many neurocognitive diseases, while its role in POCD remains obscure. Carnosine is a natural endogenous dipeptide with anti-inflammatory and neuroprotective effects. To explore the effect of carnosine on POCD and its mechanism, we established a POCD model by exploratory laparotomy in 24-month-old male Sprague-Dawley rats. We found that the administrated of carnosine notably attenuated surgery-induced NLRP3 inflammasome activation and pyroptosis in astrocytes, central inflammation, and neuronal damage in the hippocampus of aged rats. In addition, carnosine dramatically ameliorated the learning and memory deficits of surgery-induced aged rats. Then in the in vitro experiments, we stimulated primary astrocytes with lipopolysaccharide (LPS) after carnosine pretreatment. The results also showed that the application of carnosine alleviated the activation of the NLRP3 inflammasome, pyroptosis, and inflammatory response in astrocytes stimulated by LPS. Taken together, these findings suggest that carnosine improves POCD in aged rats via inhibiting NLRP3-mediated astrocytes pyroptosis and neuroinflammation.
Collapse
Affiliation(s)
- Jiahong Shen
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jiawen Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yuxin Wen
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zili Tang
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Li
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jianliang Sun
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China; Zhejiang University School of Medicine, Hangzhou, China; Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
20
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
21
|
Lou Y, Li Z, Zheng H, Yuan Z, Li W, Zhang J, Shen W, Gao Y, Ran N, Kong X, Feng S. New strategy to treat spinal cord injury: Nafamostat mesilate suppressed NLRP3-mediated pyroptosis during acute phase. Int Immunopharmacol 2024; 134:112190. [PMID: 38703569 DOI: 10.1016/j.intimp.2024.112190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
Spinal cord injury (SCI) is a devastating condition for which effective clinical treatment is currently lacking. During the acute phase of SCI, myriad pathological changes give rise to subsequent secondary injury. The results of our previous studies indicated that treating rats post-SCI with nafamostat mesilate (NM) protected the blood-spinal cord barrier (BSCB) and exerted an antiapoptotic effect. However, the optimal dosage for mice with SCI and the underlying mechanisms potentially contributing to recovery, especially during the acute phase of SCI, have not been determined. In this study, we first determined the optimal dosage of NM for mice post-SCI (5 mg/kg/day). Subsequently, our RNA-seq findings revealed that NM has the potential to inhibit pyroptosis after SCI. These findings were further substantiated by subsequent Western blot (WB) and Immunofluorescence (IF) analyses in vivo. These results indicate that NM can alleviate NLRP3 (NOD-like receptor thermal protein domain associated protein 3)-mediated pyroptosis by modulating the NF-κB signaling pathway and reducing the protein expression levels of NIMA-related kinase 7 (NEK7) and cathepsin B (CTSB). In vitro experimental results supported our in vivo findings, revealing the effectiveness of NM in suppressing pyroptosis induced by adenosine triphosphate (ATP) and lipopolysaccharide (LPS) in BV2 cells. These results underscore the potential of NM to regulate NLRP3-mediated pyroptosis following SCI. Notably, compared with other synthetic compounds, NM exhibits greater versatility, suggesting that it is a promising clinical treatment option for SCI.
Collapse
Affiliation(s)
- Yongfu Lou
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Zonghao Li
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Han Zheng
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Zhongze Yuan
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Wenxiang Li
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Jianping Zhang
- Division of Surgery and Interventional Science, University College London, London HA7 4LP, United Kingdom
| | - Wenyuan Shen
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Yiming Gao
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China
| | - Ning Ran
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China.
| | - Xiaohong Kong
- Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China.
| | - Shiqing Feng
- Department of Orthopedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University, Shandong, China.
| |
Collapse
|
22
|
Du G, Yang Z, Wen Y, Li X, Zhong W, Li Z, Zhang S, Luo E, Ding H, Li W. Heat stress induces IL-1β and IL-18 overproduction via ROS-activated NLRP3 inflammasome: implication in neuroinflammation in mice with heat stroke. Neuroreport 2024; 35:558-567. [PMID: 38687900 DOI: 10.1097/wnr.0000000000002042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Heat stroke induced cerebral damage via neuroinflammation. This study aimed to approach whether heat stress would promote NOD-like receptor protein 3 (NLRP3) inflammasome via reactive oxygen species (ROS). The mice were randomly divided into the sham group, the heat stress group, and the heat stress + TEMPOL (ROS scavenger) group. And the NLRP3 -/- mice were applied and divided into the NLRP3 -/- + sham group and the NLRP3 -/- + heat stress group. Furthermore, the BV2 cells were divided into four groups following the intervention measures: the heat stress + TEMPOL group, the heat stress + Z-VAD-FMK (caspase-1 inhibitor) group, the heat stress group, and the control group. ROS levels were examined. The expression levels of NLRP3, caspase-1, IL-1β, and IL-18 were detected by western blotting and double immunofluorescence. We found that heat stress attack induced excessive ROS in microglia and subsequently activated NLRP3 inflammasome in both mice and BV2 cells. When ROS scavenged, the expression level of NLRP3 was downregulated. Furthermore, with NLRP3 inflammasome activation, the expression levels of caspase-1, IL-1β, and IL-18 were increased. In NLRP3 -/- mice, however, the caspase-1, IL-1β, and IL-18 were significantly declined. Further experiments showed that pretreatment of caspase-1 inhibitor decreased the expression levels of IL-1β and IL-18. These results suggest that heat stress attack caused neuroinflammation via excessive ROS activating the NLRP3 inflammasome in microglia cells.
Collapse
Affiliation(s)
- Guoqiang Du
- Department of Emergency Medicine, Luoding People's Hospital, Yunfu
| | - Zixi Yang
- College of Continuing Education, Guangdong Medical University, Zhanjiang
| | - Yin Wen
- Department of Critical Care Medicine
| | - Xusheng Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou
| | | | - Zhuo Li
- Department of Critical Care Medicine
| | | | - Ensi Luo
- Department of Endocrinology, Binhaiwan Central Hospital of Dongguan, Dongguan Hospital Affiliated to Medical College of Jinan University, Dongguan, China
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou
| | - Weifeng Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou
| |
Collapse
|
23
|
Wissemann J, Heidenreich A, Zimmermann H, Engelmann J, Jansen J, Suchanek D, Westermann D, Wolf D, Stachon P, Merz J. ADP as a novel stimulus for NLRP3-inflammasome activation in mice fails to translate to humans. Purinergic Signal 2024; 20:291-302. [PMID: 37410223 PMCID: PMC11189352 DOI: 10.1007/s11302-023-09953-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023] Open
Abstract
The NLRP3-inflammasome is a cytosolic multiprotein complex that triggers an inflammatory response to certain danger signals. Recently adenosine diphosphate (ADP) was found to activate the NLRP3-inflammasome in murine macrophages via the P2Y1 receptor. Blockade of this signaling pathway reduced disease severity in a murine colitis-model. However, the role of the ADP/P2Y1-axis has not yet been studied in humans. This present study confirmed ADP-dependent NLRP3-inflammasome activation in murine macrophages, but found no evidence for a role of ADP in inflammasome activation in humans. We investigated the THP1 cell line as well as primary monocytes and further looked at macrophages. Although all cells express the three human ADP-receptors P2Y1, P2Y12 and P2Y13, independent of priming, neither increased ASC-speck formation could be detected with flow cytometry nor additional IL-1β release be found in the culture supernatant of ADP stimulated cells. We now show for the first time that the responsiveness of monocytes and macrophages to ADP as well as the regulation of its purinergic receptors is very much dependent on the species. Therefore the signaling pathway found to contribute to colitis in mice is likely not applicable to humans.
Collapse
Affiliation(s)
- Julius Wissemann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Adrian Heidenreich
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Helene Zimmermann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Juliane Engelmann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Jasper Jansen
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dymphie Suchanek
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Julian Merz
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
24
|
Zhao C, Zheng T, Wang R, Lin X, Hu Z, Zhao Z, Dai Z, Sun D. Synergistically Augmenting Cancer Immunotherapy by Physical Manipulation of Pyroptosis Induction. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:298-312. [PMID: 39398428 PMCID: PMC11466912 DOI: 10.1007/s43657-023-00140-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 10/15/2024]
Abstract
Pyroptosis is a newly recognized type of programmed cell death mediated by the gasdermin family and caspase. It is characterized by the formation of inflammasomes and the following inflammatory responses. Recent studies have elucidated the value of pyroptosis induction in cancer treatment. The inflammatory cytokines produced during pyroptosis can trigger immune responses to suppress malignancy. Physical approaches for cancer treatment, including radiotherapy, light-based techniques (photodynamic and photothermal therapy), ultrasound-based techniques (sonodynamic therapy and focused ultrasound), and electricity-based techniques (irreversible electroporation and radiofrequency ablation), are effective in clinical application. Recent studies have reported that pyroptosis is involved in the treatment process of physical approaches. Manipulating pyroptosis using physical approaches can be utilized in combating cancer, according to recent studies. Pyroptosis-triggered immunotherapy can be combined with the original anti-tumor methods to achieve a synergistic therapy and improve the therapeutic effect. Studies have also revealed that enhancing pyroptosis may increase the sensitivity of cancer cells to some physical approaches. Herein, we present a comprehensive review of the literature focusing on the associations between pyroptosis and various physical approaches for cancer and its underlying mechanisms. We also discussed the role of pyroptosis-triggered immunotherapy in the treatment process of physical manipulation.
Collapse
Affiliation(s)
- Chenyang Zhao
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| | - Tingting Zheng
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| | - Run Wang
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| | - Xiaona Lin
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| | - Zhengming Hu
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| | - Zhuofei Zhao
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, National Biomedical Imaging Centre, Peking University, Beijing, 100871 China
| | - Desheng Sun
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, 518036 Guangdong China
| |
Collapse
|
25
|
Vizuete AFK, Fróes F, Seady M, Hansen F, Ligabue-Braun R, Gonçalves CA, Souza DO. A Mechanism of Action of Metformin in the Brain: Prevention of Methylglyoxal-Induced Glutamatergic Impairment in Acute Hippocampal Slices. Mol Neurobiol 2024; 61:3223-3239. [PMID: 37980327 DOI: 10.1007/s12035-023-03774-1] [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: 05/24/2023] [Accepted: 11/05/2023] [Indexed: 11/20/2023]
Abstract
Metformin, a biguanide compound (N-1,1-dimethylbiguanide), is widely prescribed for diabetes mellitus type 2 (T2D) treatment. It also presents a plethora of properties, such as anti-oxidant, anti-inflammatory, anti-apoptosis, anti-tumorigenic, and anti-AGE formation activity. However, the precise mechanism of action of metformin in the central nervous system (CNS) needs to be clarified. Herein, we investigated the neuroprotective role of metformin in acute hippocampal slices exposed to methylglyoxal (MG), a highly reactive dicarbonyl compound and a key molecule in T2D developmental pathophysiology. Metformin protected acute hippocampal slices from MG-induced glutamatergic neurotoxicity and neuroinflammation by reducing IL-1β synthesis and secretion and RAGE protein expression. The drug also improved astrocyte function, particularly with regard to the glutamatergic system, increasing glutamate uptake. Moreover, we observed a direct effect of metformin on glutamate transporters, where the compound prevented glycation, by facilitating enzymatic phosphorylation close to Lys residues, suggesting a new neuroprotective role of metformin via PKC ζ in preventing dysfunction in glutamatergic system induced by MG.
Collapse
Affiliation(s)
- Adriana Fernanda K Vizuete
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil.
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil.
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil.
| | - Fernanda Fróes
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| | - Marina Seady
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| | - Fernanda Hansen
- Department of Nutrition, Health Sciences Center, Federal University of Santa Catarina, University Campus, Trindade, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Rodrigo Ligabue-Braun
- Department of Pharmacosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Avenida Sarmento Leite 245, Porto Alegre, 90050-130, Brazil
| | - Carlos-Alberto Gonçalves
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| | - Diogo O Souza
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
26
|
Jiang L, Wang Z, Xu T, Zhang L. When pyro(ptosis) meets palm(itoylation). Cytokine Growth Factor Rev 2024; 77:30-38. [PMID: 38472042 DOI: 10.1016/j.cytogfr.2024.03.001] [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: 02/01/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024]
Abstract
Pyroptosis, a programmed cell death process, is vital for the immune response against microbial infections and internal danger signals. Recent studies have highlighted the importance of protein palmitoylation, a modification that involves attaching palmitate to cysteine residues, in regulating key proteins involved in pyroptosis. Palmitoylation of cGAS at residue C474 by ZDHHC18 affects its enzymatic activity and DNA binding ability. Similarly, ZDHHC9 promotes cGAS activity through palmitoylation at residues C404/405. NLRP3 palmitoylation at residue C844, mediated by ZDHHC12, impacts its stability and interactions with other proteins, crucial for activating the NLRP3 inflammasome and triggering inflammation. However, the role of ZDHHC5 in NLRP3 palmitoylation remains uncertain due to conflicting findings. Palmitoylation at C88/91 is essential for STING activation and induction of type I interferons. It modulates the formation of multimeric complexes and downstream signaling pathways. GSDMD palmitoylation at C191 is necessary for pore formation and membrane translocation, while GSDME palmitoylation at C407/408 is associated with drug-induced pyroptosis. Moreover, palmitoylation of NOD1 and NOD2 influences their membrane recruitment and immune signaling pathways in response to bacterial peptidoglycans, acting as upstream regulators of pyroptosis. This review summarizes the important roles for palmitoylation in regulating the function of key pyroptosis-related proteins, shedding light on the intricate mechanisms governing immune responses and inflammation.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250013, China; Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zirui Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250013, China; Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ting Xu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250013, China; Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
27
|
Tan L, Zhang H, Ding Y, Huang Y, Sun D. CRTAC1 identified as a promising diagnosis and prognostic biomarker in lung adenocarcinoma. Sci Rep 2024; 14:11223. [PMID: 38755183 PMCID: PMC11099150 DOI: 10.1038/s41598-024-61804-x] [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: 02/01/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
CRTAC1, one of the pyroptosis-related genes, has been identified as a protective factor in certain kinds of cancer, such as gastric adenocarcinoma and bladder cancer. The study aimed to investigate the role of CRTAC1 in lung adenocarcinoma (LUAD). LUAD datasets were obtained from Gene Expression Omnibus (GEO) database and The Cancer Genome Atlas (TCGA), pyroptosis-related genes from GeneCard. Limma package used to find differentially expressed genes (DEGs), least absolute shrinkage and selection operator (LASSO) regression and weighted genes co-expression network analysis (WGCNA) to identify CRTAC1 as hub gene. CRTAC1 expression was confirmed in a real-world cohort using quantitative polymerase chain reaction (qPCR) and Western Blot (WB) analyses. Cellular experiments were conducted to investigate CRTAC1's potential oncogenic mechanisms. CRTAC1 mRNA expression was significantly lower in LUAD tissues (p < 0.05) and showed high accuracy in diagnosing LUAD. Reduced CRTAC1 expression was associated with a poor prognosis. Higher CRTAC1 expression correlated with increased immune cell infiltration. Individuals with high CRTAC1 expression showed increased drug sensitivity. Additionally, qPCR and WB analyses showed that CRTAC1 expression was lower in tumor tissue compared to adjacent normal tissue at both the RNA and protein levels. Upregulation of CRTAC1 significantly inhibited LUAD cell proliferation, invasion, and migration in cellular experiments. CRTAC1 has the potential to serve as a diagnostic and prognostic biomarker in LUAD.
Collapse
Affiliation(s)
- Lin Tan
- Tianjin Medical University Graduate School, Tianjin, China
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Han Zhang
- Tianjin Medical University Graduate School, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Yun Ding
- Tianjin Medical University Graduate School, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Yangyun Huang
- Tianjin Medical University Graduate School, Tianjin, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Daqiang Sun
- Tianjin Chest Hospital, Tianjin University, Tianjin, China.
| |
Collapse
|
28
|
Ducza L, Gaál B. The Neglected Sibling: NLRP2 Inflammasome in the Nervous System. Aging Dis 2024; 15:1006-1028. [PMID: 38722788 PMCID: PMC11081174 DOI: 10.14336/ad.2023.0926-1] [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: 08/01/2023] [Accepted: 09/26/2023] [Indexed: 05/13/2024] Open
Abstract
While classical NOD-like receptor pyrin domain containing protein 1 (NLRP1) and NLRP3 inflammasomal proteins have been extensively investigated, the contribution of NLRP2 is still ill-defined in the nervous system. Given the putative significance of NLRP2 in orchestrating neuroinflammation, further inquiry is needed to gain a better understanding of its connectome, hence its specific targeting may hold a promising therapeutic implication. Therefore, bioinformatical approach for extracting information, specifically in the context of neuropathologies, is also undoubtedly preferred. To the best of our knowledge, there is no review study selectively targeting only NLRP2. Increasing, but still fragmentary evidence should encourage researchers to thoroughly investigate this inflammasome in various animal- and human models. Taken together, herein we aimed to review the current literature focusing on the role of NLRP2 inflammasome in the nervous system and more importantly, we provide an algorithm-based protein network of human NLRP2 for elucidating potentially valuable molecular partnerships that can be the beginning of a new discourse and future therapeutic considerations.
Collapse
Affiliation(s)
- László Ducza
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary, Hungary
| | - Botond Gaál
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary, Hungary
| |
Collapse
|
29
|
Huang Y, Jiang C, Liu X, Tang W, Gui H, Sun T, Xu D, He M, Han M, Qiu H, Chen M, Huang S. Melatonin suppresses TLR4-mediated RSV infection in the central nervous cells by inhibiting NLRP3 inflammasome formation and autophagy. J Cell Mol Med 2024; 28:e18338. [PMID: 38683122 PMCID: PMC11057421 DOI: 10.1111/jcmm.18338] [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: 09/06/2023] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Respiratory syncytial virus (RSV) infects neuronal cells in the central nervous system (CNS), resulting in neurological symptoms. In the present study, we intended to explore the mechanism of RSV infection-induced neuroinflammatory injury from the perspective of the immune response and sought to identify effective protective measures against the injury. The findings showed that toll-like receptor 4 (TLR4) was activated after RSV infection in human neuronal SY5Y cells. Furthermore, TLR4 activation induced autophagy and apoptosis in neuronal cells, promoted the formation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, and increased the secretion of downstream inflammatory cytokines such as interleukin-1β (IL-1β), interleukin-18 (IL-18) and tumour necrosis factor-α (TNF-α). Interestingly, blockade of TLR4 or treatment with exogenous melatonin significantly suppressed TLR4 activation as well as TLR4-mediated apoptosis, autophagy and immune responses. Therefore, we infer that melatonin may act on the TLR4 to ameliorate RSV-induced neuronal injury, which provides a new therapeutic target for RSV infection.
Collapse
Affiliation(s)
- Yixuan Huang
- Department of EndocrinologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Chengcheng Jiang
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Xiaojie Liu
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Wei Tang
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Hongya Gui
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Tao Sun
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Doudou Xu
- Department of PediatricsThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Maozhang He
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Maozhen Han
- School of Life SciencesAnhui Medical UniversityHefeiChina
| | - Huan Qiu
- School of NursingAnhui Medical UniversityHefeiChina
| | - Mingwei Chen
- Department of EndocrinologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Shenghai Huang
- Department of Microbiology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
- School of Life SciencesAnhui Medical UniversityHefeiChina
- Department of Clinical LaboratoryAnhui Public Health Clinical Center, The First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
30
|
Shin MS, Lee Y, Cho IH, Yang HJ. Brain plasticity and ginseng. J Ginseng Res 2024; 48:286-297. [PMID: 38707640 PMCID: PMC11069001 DOI: 10.1016/j.jgr.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/10/2024] [Accepted: 03/21/2024] [Indexed: 05/07/2024] Open
Abstract
Brain plasticity refers to the brain's ability to modify its structure, accompanied by its functional changes. It is influenced by learning, experiences, and dietary factors, even in later life. Accumulated researches have indicated that ginseng may protect the brain and enhance its function in pathological conditions. There is a compelling need for a more comprehensive understanding of ginseng's role in the physiological condition because many individuals without specific diseases seek to improve their health by incorporating ginseng into their routines. This review aims to deepen our understanding of how ginseng affects brain plasticity of people undergoing normal aging process. We provided a summary of studies that reported the impact of ginseng on brain plasticity and related factors in human clinical studies. Furthermore, we explored researches focused on the molecular mechanisms underpinning the influence of ginseng on brain plasticity and factors contributing to brain plasticity. Evidences indicate that ginseng has the potential to enhance brain plasticity in the context of normal aging by mediating both central and peripheral systems, thereby expecting to improve age-related declines in brain function. Moreover, given modern western diet can damage neuroplasticity in the long term, ginseng can be a beneficial supplement for better brain health.
Collapse
Affiliation(s)
- Myoung-Sook Shin
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - YoungJoo Lee
- Department of Integrative Bioscience and Biotechnology, College of Life Science, Sejong University, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun-Jeong Yang
- Department of Integrative Bioscience, University of Brain Education, Cheonan, Republic of Korea
- Department of Integrative Healthcare, University of Brain Education, Cheonan, Republic of Korea
- Korea Institute of Brain Science, Seoul, Republic of Korea
| |
Collapse
|
31
|
Sun X, Lu Y, Pang Q, Luo B, Jiang Q. Tooth loss impairs cognitive function in SAMP8 mice via the NLRP3/Caspase-1 pathway. Oral Dis 2024; 30:2746-2755. [PMID: 37357357 DOI: 10.1111/odi.14646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/13/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2023]
Abstract
OBJECTIVE Loss of occlusal support due to tooth loss has been indicated as one of the risk factors for Alzheimer's disease. This study aimed to investigate the relationship between tooth loss and cognitive dysfunction and illustrate the role of neuroinflammation in advancing Alzheimer's disease. MATERIALS AND METHODS Male 5-month-old senescence-accelerated mouse strain P8 (SAMP8) mice were divided into three groups (n = 7): the C (control), S (sham-operated), and TL (tooth loss) groups. The Morris water maze (MWM) test was performed to assess spatial memory. Additionally, histopathological and molecular assessments of hippocampal tissues were performed. RESULTS The TL groups exhibited impaired spatial memory in the water maze. Tooth loss induced higher protein expression levels of the neuroinflammation cytokine interleukin-1β (IL-1β) in the hippocampus than in the S and C groups. Tooth loss activated the NLRP3 inflammasome and increased the expression of Caspase-1 in the hippocampus. CONCLUSIONS The findings indicated that tooth loss impairs cognitive function in SAMP8 mice and is closely related to the activation of NLRP3/Caspase-1 in the hippocampus.
Collapse
Affiliation(s)
- Xu Sun
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yunping Lu
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Qian Pang
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Bin Luo
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Qingsong Jiang
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Allen J, Isaza-Correa J, Kelly L, Melo A, Mahony A, McDonald D, Molloy EJ. Severe neurological impairment and immune function: altered neutrophils, monocytes, T lymphocytes, and inflammasome activation. Pediatr Res 2024; 95:1611-1616. [PMID: 38233513 PMCID: PMC11126379 DOI: 10.1038/s41390-024-03023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/18/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Infections cause significant morbidity and mortality in children with Severe Neurological Impairment (SNI). Alterations in immune cell numbers and function in children with neurodisability have been reported. We aimed to characterise neutrophil, monocyte and lymphocyte proportions and activation, at baseline and in response to stimulation with lipopolysaccharide, in children with SNI compared to healthy controls. METHODS Whole blood samples of children with SNI and controls were incubated in the presence or absence of lipopolysaccharide (10 ng/ml). Monocyte and neutrophil function (Cluster of Differentiation (CD)11b, (TLR)-4 and CD66b expression) and lymphocytes were assessed by flow cytometry. Expression of genes involved in the inflammasome (NLR Family Pyrin Domain Containing(NLRP)-3, Apoptosis-Associated Speck-like protein (ASC) and Interleukin(IL)1β) were assessed by PCR. RESULTS Monocytes and CD8+ T cells were lower in children with SNI (n = 14). CD66b, was hyporesponsive and monocyte TLR4 was hyperresponsive to lipopolysaccharide in children with SNI compared to controls (n = 14). NLRP3 expression was higher at baseline and IL1β expression was not upregulated in response to lipopolysaccharide in children with SNI in contrast to controls. CONCLUSION We have found significant differences in immune regulation in children with SNI compared to controls which may provide a useful therapeutic target in the future. IMPACT Children with SNI have reduced monocyte and CD8+ T cells. Neutrophils and monocytes in children with SNI show altered markers of activation in response to lipopolysaccharide. Expression of NLRP3 at the RNA level was higher at baseline in children with SNI. This study adds to the existing literature that children with neurological impairment have altered inflammatory and immune cell responses. This may provide a useful therapeutic target to reduce infection-related morbidity and mortality, and tertiary neurological injury in the future.
Collapse
Affiliation(s)
- John Allen
- Discipline of Paediatrics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute (TTMI) Trinity College Dublin, Dublin, Ireland
- Department of Neurodisability, Children's Health Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Johana Isaza-Correa
- Discipline of Paediatrics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute (TTMI) Trinity College Dublin, Dublin, Ireland
| | - Lynne Kelly
- Discipline of Paediatrics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute (TTMI) Trinity College Dublin, Dublin, Ireland
| | - Ashanty Melo
- Discipline of Paediatrics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute (TTMI) Trinity College Dublin, Dublin, Ireland
| | - Aoife Mahony
- Department of Neurodisability, Children's Health Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Denise McDonald
- Discipline of Paediatrics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, the University of Dublin, Dublin, Ireland
- Department of Neurodisability, Children's Health Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Eleanor J Molloy
- Discipline of Paediatrics, School of Medicine, Trinity College Dublin, the University of Dublin, Dublin, Ireland.
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, the University of Dublin, Dublin, Ireland.
- Trinity Translational Medicine Institute (TTMI) Trinity College Dublin, Dublin, Ireland.
- Department of Neurodisability, Children's Health Ireland (CHI) at Tallaght, Dublin, Ireland.
- Department of Neonatology, CHI at Crumlin, Dublin, Ireland.
- Coombe Hospital, Dublin, Ireland.
| |
Collapse
|
33
|
Hedley KE, Cuskelly A, Callister RJ, Horvat JC, Hodgson DM, Tadros MA. The medulla oblongata shows a sex-specific inflammatory response to systemic neonatal lipopolysaccharide. J Neuroimmunol 2024; 389:578316. [PMID: 38394966 DOI: 10.1016/j.jneuroim.2024.578316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Early life inflammation has been linked to long-term modulation of behavioural outcomes due to the central nervous system, but it is now becoming apparent it is also linked to dysfunction of visceral physiology. The medulla oblongata contains a number of nuclei critical for homeostasis, therefore we utilised the well-established model of neonatal lipopolysaccharide (LPS) exposure to examine the immediate and long-term impacts of systemic inflammation on the medulla oblongata. Wistar rats were injected with LPS or saline on postnatal days 3 and 5, with tissues collected on postnatal days 7 or 90 in order to assess expression of inflammatory mediators and microglial morphology in autonomic regions of the medulla oblongata. We observed a distinct sex-specific response of all measured inflammatory mediators at both ages, as well as significant neonatal sex differences in inflammatory mediators within saline groups. At both ages, microglial morphology had significant changes in branch length and soma size in a sex-specific manner in response to LPS exposure. This data not only highlights the strong sex-specific response of neonates to LPS administration, but also the significant life-long impact on the medulla oblongata and the potential altered control of visceral organs.
Collapse
Affiliation(s)
- Kateleen E Hedley
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Annalisa Cuskelly
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Psychological Sciences, University of Newcastle, NSW, Australia; School of Education, University of Newcastle, NSW, Australia
| | - Robert J Callister
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jay C Horvat
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Deborah M Hodgson
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Psychological Sciences, University of Newcastle, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
34
|
Thal DR, Gawor K, Moonen S. Regulated cell death and its role in Alzheimer's disease and amyotrophic lateral sclerosis. Acta Neuropathol 2024; 147:69. [PMID: 38583129 DOI: 10.1007/s00401-024-02722-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 04/08/2024]
Abstract
Despite considerable research efforts, it is still not clear which mechanisms underlie neuronal cell death in neurodegenerative diseases. During the last 20 years, multiple pathways have been identified that can execute regulated cell death (RCD). Among these RCD pathways, apoptosis, necroptosis, pyroptosis, ferroptosis, autophagy-related cell death, and lysosome-dependent cell death have been intensively investigated. Although RCD consists of numerous individual pathways, multiple common proteins have been identified that allow shifting from one cell death pathway to another. Another layer of complexity is added by mechanisms such as the endosomal machinery, able to regulate the activation of some RCD pathways, preventing cell death. In addition, restricted axonal degeneration and synaptic pruning can occur as a result of RCD activation without loss of the cell body. RCD plays a complex role in neurodegenerative processes, varying across different disorders. It has been shown that RCD is differentially involved in Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS), among the most common neurodegenerative diseases. In AD, neuronal loss is associated with the activation of not only necroptosis, but also pyroptosis. In ALS, on the other hand, motor neuron death is not linked to canonical necroptosis, whereas pyroptosis pathway activation is seen in white matter microglia. Despite these differences in the activation of RCD pathways in AD and ALS, the accumulation of protein aggregates immunoreactive for p62/SQSTM1 (sequestosome 1) is a common event in both diseases and many other neurodegenerative disorders. In this review, we describe the major RCD pathways with clear activation in AD and ALS, the main interactions between these pathways, as well as their differential and similar involvement in these disorders. Finally, we will discuss targeting RCD as an innovative therapeutic concept for neurodegenerative diseases, such as AD and ALS. Considering that the execution of RCD or "cellular suicide" represents the final stage in neurodegeneration, it seems crucial to prevent neuronal death in patients by targeting RCD. This would offer valuable time to address upstream events in the pathological cascade by keeping the neurons alive.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute (LBI), KU-Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Klara Gawor
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute (LBI), KU-Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Sebastiaan Moonen
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute (LBI), KU-Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven Brain Institute (LBI), Leuven, Belgium
- Center for Brain & Disease Research, VIB, Leuven, Belgium
| |
Collapse
|
35
|
Argandona Lopez C, Brown AM. Microglial- neuronal crosstalk in chronic viral infection through mTOR, SPP1/OPN and inflammasome pathway signaling. Front Immunol 2024; 15:1368465. [PMID: 38646526 PMCID: PMC11032048 DOI: 10.3389/fimmu.2024.1368465] [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: 01/10/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
HIV-infection of microglia and macrophages (MMs) induces neuronal injury and chronic release of inflammatory stimuli through direct and indirect molecular pathways. A large percentage of people with HIV-associated neurologic and psychiatric co-morbidities have high levels of circulating inflammatory molecules. Microglia, given their susceptibility to HIV infection and long-lived nature, are reservoirs for persistent infection. MMs and neurons possess the molecular machinery to detect pathogen nucleic acids and proteins to activate innate immune signals. Full activation of inflammasome assembly and expression of IL-1β requires a priming event and a second signal. Many studies have demonstrated that HIV infection alone can activate inflammasome activity. Interestingly, secreted phosphoprotein-1 (SPP1/OPN) expression is highly upregulated in the CNS of people infected with HIV and neurologic dysfunction. Interestingly, all evidence thus far suggests a protective function of SPP1 signaling through mammalian target of rapamycin (mTORC1/2) pathway function to counter HIV-neuronal injury. Moreover, HIV-infected mice knocked down for SPP1 show by neuroimaging, increased neuroinflammation compared to controls. This suggests that SPP1 uses unique regulatory mechanisms to control the level of inflammatory signaling. In this mini review, we discuss the known and yet-to-be discovered biological links between SPP1-mediated stimulation of mTOR and inflammasome activity. Additional new mechanistic insights from studies in relevant experimental models will provide a greater understanding of crosstalk between microglia and neurons in the regulation of CNS homeostasis.
Collapse
Affiliation(s)
- Catalina Argandona Lopez
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amanda M. Brown
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Neuroimmunology, Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
36
|
Yi H, Zhu B, Zheng C, Ying Z, Cheng M. CXCL13/CXCR5 promote chronic postsurgical pain and astrocyte activation in rats by targeting NLRP3. Neuroreport 2024; 35:406-412. [PMID: 38526919 DOI: 10.1097/wnr.0000000000002023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Chronic postsurgical pain (CPSP) with high incidence negatively impacts the quality of life. X-C motif chemokine 13 (CXCL13) has been associated with postsurgery inflammation and exacerbates neuropathic pain in patients with CPSP. This study was aimed to illustrate the relationship between CXCL13 and nod-like receptor protein-3 (NLRP3), which is also involved in CPSP. A CPSP model was constructed by skin/muscle incision and retraction (SMIR) in right medial thigh, and the rats were divided into three groups: Sham, SMIR, and SMIR + anti-CXCL13 (intrathecally injected with anti-CXCL13 antibody). Then, the paw withdrawal threshold (PWT) score of rats was recorded. Primary rat astrocytes were isolated and treated with recombinant protein CXCL13 with or without NLRP3 inhibitor INF39. The expressions of CXCL13, CXCR5, IL-1β, IL-18, GFAP, NLRP3, and Caspase-1 p20 were detected by real-time quantitative reverse transcription PCR, western blot, ELISA, immunocytochemistry, and immunofluorescence analyses. The anti-CXCL13 antibody alleviated SMIR-induced decreased PWT and increased expression of GFAP, CXCL13, CXCR5, NLRP3, and Caspase-1 p20 in spinal cord tissues. The production of IL-1β, IL-18, and expression of CXCL13, CXCR5, GFAP, NLRP3, and Caspase-1 p20 were increased in recombinant protein CXCL13-treated primary rat astrocytes in a dose-dependent manner. Treatment with NLRP3 inhibitor INF39 inhibited the function of recombinant protein CXCL13 in primary rat astrocytes. The CXCL13/CXCR5 signaling could promote neuropathic pain, astrocytes activation, and NLRP3 inflammasome activation in CPSP model rats by targeting NLRP3. NLRP3 may be a potential target for the management of CPSP.
Collapse
Affiliation(s)
- Hongda Yi
- Department of Anesthesiology, Hangzhou Women's Hospital, Hangzhou, China
| | | | | | | | | |
Collapse
|
37
|
Oladapo A, Jackson T, Menolascino J, Periyasamy P. Role of pyroptosis in the pathogenesis of various neurological diseases. Brain Behav Immun 2024; 117:428-446. [PMID: 38336022 PMCID: PMC10911058 DOI: 10.1016/j.bbi.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/22/2023] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Pyroptosis, an inflammatory programmed cell death process, has recently garnered significant attention due to its pivotal role in various neurological diseases. This review delves into the intricate molecular signaling pathways governing pyroptosis, encompassing both caspase-1 dependent and caspase-1 independent routes, while emphasizing the critical role played by the inflammasome machinery in initiating cell death. Notably, we explore the Nucleotide-binding domain leucine-rich repeat (NLR) containing protein family, the Absent in melanoma 2-like receptor family, and the Pyrin receptor family as essential activators of pyroptosis. Additionally, we comprehensively examine the Gasdermin family, renowned for their role as executioner proteins in pyroptosis. Central to our review is the interplay between pyroptosis and various central nervous system (CNS) cell types, including astrocytes, microglia, neurons, and the blood-brain barrier (BBB). Pyroptosis emerges as a significant factor in the pathophysiology of each cell type, highlighting its far-reaching impact on neurological diseases. This review also thoroughly addresses the involvement of pyroptosis in specific neurological conditions, such as HIV infection, drug abuse-mediated pathologies, Alzheimer's disease, and Parkinson's disease. These discussions illuminate the intricate connections between pyroptosis, chronic inflammation, and cell death in the development of these disorders. We also conducted a comparative analysis, contrasting pyroptosis with other cell death mechanisms, thereby shedding light on their unique aspects. This approach helps clarify the distinct contributions of pyroptosis to neuroinflammatory processes. In conclusion, this review offers a comprehensive exploration of the role of pyroptosis in various neurological diseases, emphasizing its multifaceted molecular mechanisms within various CNS cell types. By elucidating the link between pyroptosis and chronic inflammation in the context of neurodegenerative disorders and infections, it provides valuable insights into potential therapeutic targets for mitigating these conditions.
Collapse
Affiliation(s)
- Abiola Oladapo
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Thomas Jackson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Jueliet Menolascino
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
38
|
Hou B, Yin J, Liu S, Guo J, Zhang B, Zhang Z, Yang L, Tan X, Long Y, Feng S, Zhou J, Wu Y, Wang X, Han S, Wang Z, He X. Inhibiting the NLRP3 Inflammasome with MCC950 Alleviates Neurological Impairment in the Brain of EAE Mice. Mol Neurobiol 2024; 61:1318-1330. [PMID: 37702910 PMCID: PMC10896958 DOI: 10.1007/s12035-023-03618-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by demyelination and neuronal damage. Experimental autoimmune encephalomyelitis (EAE) mice are used to model the disease progression of MS and mirror MS-like pathology. Previous researches have confirmed that inhibition of NLRP3 inflammasome significantly alleviated the severity of EAE mice and the demyelination of spinal cord, but its effect on neuronal damage and oligodendrocyte loss in the brain remains unclear. In this study, female C57BL/6 mice were immunized with MOG35-55 and PTX to establish experimental autoimmune encephalomyelitis (EAE) model. MCC950, a selective NLRP3 inflammasome inhibitor, was used to investigate the effect of NLRP3 inflammasome on the pathological changes and glial cell activation in the brain of EAE mice by immunohistochemistry. Our results demonstrated that MCC950 ameliorated the neuronal damage, demyelination, and oligodendrocyte loss in the brain of EAE mice. This protective effect of MCC950 may be attributed to its ability to suppress the activation of glial cells and prevents microglia polarization to M1 phenotype. Our work indicates that inhibition of NLRP3 inflammasome has the therapeutic effects of neuroprotection through immunomodulation and is a promising therapeutic strategy for MS.
Collapse
Affiliation(s)
- Baohua Hou
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
- Central Laboratory, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, China
| | - Jun Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Shuyan Liu
- Department of Endocrinology, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Jincheng Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital), Jiaozuo, 454000, China
| | - Baobao Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Zhenzhen Zhang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Lanping Yang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Xiying Tan
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Yijiao Long
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Sijie Feng
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China
| | - Jingchun Zhou
- Beijing Bencaoyuan Pharmaceutical Co, Ltd, Beijing, 102629, China
| | - Yifan Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Xueyang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Song Han
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China
| | - Zhenhui Wang
- College of Medicine, Henan Polytechnic University, Jiaozuo, 454000, China.
| | - Xiaohua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
39
|
Kodi T, Sankhe R, Gopinathan A, Nandakumar K, Kishore A. New Insights on NLRP3 Inflammasome: Mechanisms of Activation, Inhibition, and Epigenetic Regulation. J Neuroimmune Pharmacol 2024; 19:7. [PMID: 38421496 PMCID: PMC10904444 DOI: 10.1007/s11481-024-10101-5] [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: 03/07/2023] [Accepted: 11/06/2023] [Indexed: 03/02/2024]
Abstract
Inflammasomes are important modulators of inflammation. Dysregulation of inflammasomes can enhance vulnerability to conditions such as neurodegenerative diseases, autoinflammatory diseases, and metabolic disorders. Among various inflammasomes, Nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) is the best-characterized inflammasome related to inflammatory and neurodegenerative diseases. NLRP3 is an intracellular sensor that recognizes pathogen-associated molecular patterns and damage-associated patterns resulting in the assembly and activation of NLRP3 inflammasome. The NLRP3 inflammasome includes sensor NLRP3, adaptor apoptosis-associated speck-like protein (ASC), and effector cysteine protease procaspase-1 that plays an imperative role in caspase-1 stimulation which further initiates a secondary inflammatory response. Regulation of NLRP3 inflammasome ameliorates NLRP3-mediated diseases. Much effort has been invested in studying the activation, and exploration of specific inhibitors and epigenetic mechanisms controlling NLRP3 inflammasome. This review gives an overview of the established NLRP3 inflammasome assembly, its brief molecular mechanistic activations as well as a current update on specific and non-specific NLRP3 inhibitors that could be used in NLRP3-mediated diseases. We also focused on the recently discovered epigenetic mechanisms mediated by DNA methylation, histone alterations, and microRNAs in regulating the activation and expression of NLRP3 inflammasome, which has resulted in a novel method of gaining insight into the mechanisms that modulate NLRP3 inflammasome activity and introducing potential therapeutic strategies for CNS disorders.
Collapse
Affiliation(s)
- Triveni Kodi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Adarsh Gopinathan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
40
|
Stergiou IE, Tsironis C, Papadakos SP, Tsitsilonis OE, Dimopoulos MA, Theocharis S. Unraveling the Role of the NLRP3 Inflammasome in Lymphoma: Implications in Pathogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:2369. [PMID: 38397043 PMCID: PMC10889189 DOI: 10.3390/ijms25042369] [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: 01/03/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammasomes are multimeric protein complexes, sensors of intracellular danger signals, and crucial components of the innate immune system, with the NLRP3 inflammasome being the best characterized among them. The increasing scientific interest in the mechanisms interconnecting inflammation and tumorigenesis has led to the study of the NLRP3 inflammasome in the setting of various neoplasms. Despite a plethora of data regarding solid tumors, NLRP3 inflammasome's implication in the pathogenesis of hematological malignancies only recently gained attention. In this review, we investigate its role in normal lymphopoiesis and lymphomagenesis. Considering that lymphomas comprise a heterogeneous group of hematologic neoplasms, both tumor-promoting and tumor-suppressing properties were attributed to the NLRP3 inflammasome, affecting neoplastic cells and immune cells in the tumor microenvironment. NLRP3 inflammasome-related proteins were associated with disease characteristics, response to treatment, and prognosis. Few studies assess the efficacy of NLRP3 inflammasome therapeutic targeting with encouraging results, though most are still at the preclinical level. Further understanding of the mechanisms regulating NLRP3 inflammasome activation during lymphoma development and progression can contribute to the investigation of novel treatment approaches to cover unmet needs in lymphoma therapeutics.
Collapse
Affiliation(s)
- Ioanna E. Stergiou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (C.T.)
| | - Christos Tsironis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (C.T.)
| | - Stavros P. Papadakos
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece;
| | - Ourania E. Tsitsilonis
- Flow Cytometry Unit, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15784 Athens, Greece;
| | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, 11528 Athens, Greece;
| | - Stamatios Theocharis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece;
| |
Collapse
|
41
|
Zhou Y, Yang X, Zhu L. A novel Nlrp3 knock-in mouse model with hyperactive inflammasome in development of lethal inflammation. Clin Exp Immunol 2024; 215:202-214. [PMID: 37594231 PMCID: PMC10847811 DOI: 10.1093/cei/uxad097] [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: 02/22/2023] [Revised: 06/23/2023] [Accepted: 08/11/2023] [Indexed: 08/19/2023] Open
Abstract
NOD-like receptor family, pyrin domain-containing 3 (NLRP3) is a central protein contributing to human inflammatory disorders, including cryopyrin-associated periodic syndrome and sepsis. However, the molecular mechanisms and functions of NLRP3 activation in various diseases remain unknown. Here, we generated gain-of-function knock-in mice associated with Muckle-Wells syndromes using the Cre-LoxP system allowing for the constitutive T346M mutation of NLRP3 to be globally expressed in all cells under the control of tamoxifen. The mice were treated with tamoxifen for 4 days before determining their genotype by PCR and sequence analysis. In vitro, we found that bone marrow-derived macrophage from homozygous T346M mutation mice displayed a robust ability to produce IL-1β in response to lipopolysaccharide exposure. Moreover, ASC specks and oligomerization were observed in the homozygous mutant bone marrow-derived macrophages in the presence of lipopolysaccharides alone. Mechanistically, K+ and Ca2+ depletion and mitochondrial depolarization contribute to the hyperactivation of mutant NLRP3. In vivo, homozygous mice carrying the T346M mutation exhibit weight loss and mild inflammation in the resting state. In the lipopolysaccharide-mediated sepsis model, homozygous mutant mice exhibited higher mortality and increased serum circulating cytokine levels, accompanied by serious liver injury. Furthermore, an increase in myeloid cells in the spleen has been suggested to be a risk factor for inducing sepsis sensitivity. Altogether, we describe a cryopyrin-associated syndrome animal model with the T346M mutation of NLRP3 and suggest that the hyperactivated inflammasome aggregated by the mutant NLRP3 lowers the inflammatory response threshold both in vitro and in vivo.
Collapse
Affiliation(s)
- Yongting Zhou
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College; Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Xiyue Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College; Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Lei Zhu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College; Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing, PR China
| |
Collapse
|
42
|
Wenfei Z, Xiang T, Chen C, Yang T, Yun T, Zhibiao C, Ge Z. Isoliquiritigenin attenuates neuroinflammation after subarachnoid hemorrhage through inhibition of NF-κB-mediated NLRP3 inflammasome activation. Chem Biol Drug Des 2024; 103:e14436. [PMID: 38395608 DOI: 10.1111/cbdd.14436] [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: 11/02/2023] [Revised: 12/04/2023] [Accepted: 12/15/2023] [Indexed: 02/25/2024]
Abstract
Neuroinflammation contributes to neurological dysfunction in the patients who suffer from subarachnoid hemorrhage (SAH). Isoliquiritigenin (ISL) is a bioactive component extracted from Genus Glycyrrhiza. This work is to investigate whether ISL ameliorates neuroinflammation after SAH. In this study, intravascular perforation of male Sprague-Dawley rats was used to establish a SAH model. ISL was administered by intraperitoneal injection 6 h after SAH in rats. The mortality, SAH grade, neurological score, brain water content, and blood-brain barrier (BBB) permeability were examined at 24 h after the treatment. Expressions of tumor necrosis factor-α, interleukin-6, Iba-1, and MPO were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Besides, the expression levels of NF-κB p65 and NLRP3, ASC, caspase-1, IL-1β, and IL-18 were analyzed by western blot. The experimental data suggested that ISL treatment could ameliorate neurological impairment, attenuate brain edema, and ameliorate BBB injury after SAH in rats. ISL treatment repressed the expression of proinflammatory cytokines TNF-α and IL-6, and meanwhile inhibited the expression of Iba-1 and MPO. ISL also repressed NF-κB p65 expression as well as the transport from the cytoplasm to the nucleus. In addition, ISL significantly suppressed the expression levels of NLR family pyrin domain containing 3 (NLRP3), ASC, caspase-1, IL-1β, and IL-18. These findings suggest that ISL inactivates NLRP3 pathway by inhibiting NF-κB p65 translocation, thereby repressing the neuroinflammation after SAH, and it is a potential drug for the treatment of SAH.
Collapse
Affiliation(s)
- Zhang Wenfei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Xiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of Orthodontics, Wuhan First Stomatological Hospital, Wuhan, China
| | - Tao Yang
- Department of Nursing, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Yun
- Department of Stomatology, Wuhan Central Hospital, Wuhan, China
| | - Chen Zhibiao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhang Ge
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
43
|
Ciubuc-Batcu MT, Stapelberg NJC, Headrick JP, Renshaw GMC. A mitochondrial nexus in major depressive disorder: Integration with the psycho-immune-neuroendocrine network. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166920. [PMID: 37913835 DOI: 10.1016/j.bbadis.2023.166920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023]
Abstract
Nervous system processes, including cognition and affective state, fundamentally rely on mitochondria. Impaired mitochondrial function is evident in major depressive disorder (MDD), reflecting cumulative detrimental influences of both extrinsic and intrinsic stressors, genetic predisposition, and mutation. Glucocorticoid 'stress' pathways converge on mitochondria; oxidative and nitrosative stresses in MDD are largely mitochondrial in origin; both initiate cascades promoting mitochondrial DNA (mtDNA) damage with disruptions to mitochondrial biogenesis and tryptophan catabolism. Mitochondrial dysfunction facilitates proinflammatory dysbiosis while directly triggering immuno-inflammatory activation via released mtDNA, mitochondrial lipids and mitochondria associated membranes (MAMs), further disrupting mitochondrial function and mitochondrial quality control, promoting the accumulation of abnormal mitochondria (confirmed in autopsy studies). Established and putative mechanisms highlight a mitochondrial nexus within the psycho-immune neuroendocrine (PINE) network implicated in MDD. Whether lowering neuronal resilience and thresholds for disease, or linking mechanistic nodes within the MDD pathogenic network, impaired mitochondrial function emerges as an important risk, a functional biomarker, providing a therapeutic target in MDD. Several treatment modalities have been demonstrated to reset mitochondrial function, which could benefit those with MDD.
Collapse
Affiliation(s)
- M T Ciubuc-Batcu
- Griffith University School of Medicine and Dentistry, Australia; Gold Coast Health, Queensland, Australia
| | - N J C Stapelberg
- Bond University Faculty of Health Sciences and Medicine, Australia; Gold Coast Health, Queensland, Australia
| | - J P Headrick
- Griffith University School of Pharmacy and Medical Science, Australia
| | - G M C Renshaw
- Hypoxia and Ischemia Research Unit, Griffith University, School of Health Sciences and Social Work, Australia.
| |
Collapse
|
44
|
Choi BJ, Park MH, Jin HK, Bae JS. Acid sphingomyelinase as a pathological and therapeutic target in neurological disorders: focus on Alzheimer's disease. Exp Mol Med 2024; 56:301-310. [PMID: 38337058 PMCID: PMC10907607 DOI: 10.1038/s12276-024-01176-4] [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: 08/08/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 02/12/2024] Open
Abstract
Over the past decade, numerous studies have highlighted the importance of acid sphingomyelinase (ASM) in disease treatment in humans. This enzyme functions primarily to generate ceramide, maintain the cellular membrane, and regulate cellular function. However, in the blood and brain of patients with neurological disorders, including major depression, ischemic stroke, amyotrophic lateral sclerosis, multiple sclerosis, and Alzheimer's disease (AD), elevated ASM levels significantly suggest disease onset or progression. In these diseases, increased ASM is profoundly involved in neuronal death, abnormal autophagy, neuroinflammation, blood-brain barrier disruption, hippocampal neurogenesis loss, and immune cell dysfunction. Moreover, genetic and pharmacological inhibition of ASM can prevent or ameliorate various diseases. The therapeutic effects of ASM inhibition have prompted the urgent need to develop ASM inhibitors, and several ASM inhibitors have been identified. In this review, we summarize the current knowledge on the critical roles and mechanisms of ASM in brain cells and blood that are associated with different neuropathological features, especially those observed in AD. Furthermore, we elucidate the potential possibility and limitations of existing ASM-targeting drugs according to experimental studies in neurological disorder mouse models.
Collapse
Affiliation(s)
- Byung Jo Choi
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Min Hee Park
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Hee Kyung Jin
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, South Korea
| | - Jae-Sung Bae
- KNU Alzheimer's Disease Research Institute, Kyungpook National University, Daegu, 41566, South Korea.
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea.
| |
Collapse
|
45
|
Russo C, Lombardo GE, Bruschetta G, Rapisarda A, Maugeri A, Navarra M. Bergamot Byproducts: A Sustainable Source to Counteract Inflammation. Nutrients 2024; 16:259. [PMID: 38257152 PMCID: PMC10819577 DOI: 10.3390/nu16020259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Chronic inflammation is the result of an acute inflammatory response that fails to eliminate the pathogenic agent or heal the tissue injury. The consequence of this failure lays the foundations to the onset of several chronic ailments, including skin disorders, respiratory and neurodegenerative diseases, metabolic syndrome, and, eventually, cancer. In this context, the long-term use of synthetic anti-inflammatory drugs to treat chronic illnesses cannot be tolerated by patients owing to the severe side effects. Based on this, the need for novel agents endowed with anti-inflammatory effects prompted to search potential candidates also within the plant kingdom, being recognized as a source of molecules currently employed in several therapeutical areas. Indeed, the ever-growing evidence on the anti-inflammatory properties of dietary polyphenols traced the route towards the study of flavonoid-rich sources, such as Citrus bergamia (bergamot) and its derivatives. Interestingly, the recent paradigm of the circular economy has promoted the valorization of Citrus fruit waste and, in regard to bergamot, it brought to light new evidence corroborating the anti-inflammatory potential of bergamot byproducts, thus increasing the scientific knowledge in this field. Therefore, this review aims to gather the latest literature supporting the beneficial role of both bergamot derivatives and waste products in different models of inflammatory-based diseases, thus highlighting the great potentiality of a waste re-evaluation perspective.
Collapse
Affiliation(s)
- Caterina Russo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (C.R.); (G.E.L.); (A.R.); (M.N.)
| | - Giovanni Enrico Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (C.R.); (G.E.L.); (A.R.); (M.N.)
| | - Giuseppe Bruschetta
- Department of Veterinary Sciences, University of Messina, Viale G. Palatucci, 98168 Messina, Italy;
| | - Antonio Rapisarda
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (C.R.); (G.E.L.); (A.R.); (M.N.)
| | - Alessandro Maugeri
- Department of Veterinary Sciences, University of Messina, Viale G. Palatucci, 98168 Messina, Italy;
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (C.R.); (G.E.L.); (A.R.); (M.N.)
| |
Collapse
|
46
|
Luo G, Chen L, Chen M, Mao L, Zeng Q, Zou Y, Xue J, Liu P, Wu Q, Yang S, Liu M. Hirudin inhibit the formation of NLRP3 inflammasome in cardiomyocytes via suppressing oxidative stress and activating mitophagy. Heliyon 2024; 10:e23077. [PMID: 38163129 PMCID: PMC10754874 DOI: 10.1016/j.heliyon.2023.e23077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Context Cardiomyocyte hypertrophy due to hemodynamic overload eventually leads to heart failure. Hirudin has been widely used in the treatment of cardiovascular diseases and NLRP3 inflammasome was proven to induce cardiomyocyte pyroptosis. However, the mechanism by which it inhibits cardiomyocyte hypertrophy remains unclear. Objective To explore the mechanism of hirudin inhibiting cardiomyocyte hypertrophy based on NLRP3 inflammasome activation and mitophagy. Materials & methods 1 μM AngII was used for cardiac hypertrophy modeling in H9C2 cells, and cell viability was quantified by CCK-8 assay to screen the appropriate action concentrations of hirudin. After that, we cultured AngII induced-H9C2 cells for 24 h with 0, 0.3, 0.6, and 1.2 mM hirudin, respectively. Next, we marked H9C2 cells with phalloidine and observed them using fluorescence microscope. IL-1β, IL-18, IL-6, TNF-α, ANP, BNP, β-MHC, and mtDNA were analyzed by qRT-PCR; ROS were quantified by Flow cytometry; SOD, MDA, and GSH-Px were detected by ELISA; and proteins including NLRP3, ASC, caspase-1, pro-caspase-1, IL-1β, IL-18, PINK-1, Parkin, beclin-1, LC3-Ⅰ, LC3-Ⅱ, p62, were quantified by western blotting. Results It was discovered that hirudin reduced the superficial area of AngII-induced H9C2 cells and inhibited the AngII-induced up-regulation of ANP, BNP, and β-MHC. Besides, hirudin down-regulated the expressions of NLRP3 inflammasome-related cytokines, containing IL-1β, IL-18, IL-6, TNF-α. It also down-regulated the expression of mtDNA and ROS, decreased the expression levels of NLRP3 inflammasome activation related proteins, including NLRP3, ASC, caspase-1, pro-caspase-1, IL-1β, IL-18; and increased the expressions of PINK-1, Parkin, beclin-1, LC3-Ⅱ/LC3-Ⅰ, p62 in AngII-induced H9C2 cells. Discussion Hirudin promoted the process of mitophagy, inhibited the development of inflammation and oxidative stress, and inhibited the activation of the NLRP3 inflammasome and the PINK-1/Parkin pathway. Conclusion Hirudin has the activity to suppress cardiac hypertrophy may benefit from the inhibition of NLRP3 inflammasome and activating of PINK-1/Parkin related-mitophagy.
Collapse
Affiliation(s)
- Gang Luo
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Li Chen
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Mingtai Chen
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linshen Mao
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Qihu Zeng
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Yuan Zou
- School of Integrated Traditional and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinyi Xue
- School of Integrated Traditional and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| | - Mengnan Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| |
Collapse
|
47
|
Meng Y, Sun J, Zhang G. Pick fecal microbiota transplantation to enhance therapy for major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110860. [PMID: 37678703 DOI: 10.1016/j.pnpbp.2023.110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/23/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
In recent years, fecal microbiota transplantation (FMT) has emerged as a promising therapy for major depressive disorder (MDD). The goal of the operation is to restore a healthy gut microbiota by introducing feces from a healthy donor into the recipient's digestive system. The brain-gut axis is thought to have a significant role in regulating mood, behavior, and cognition, which supports the use of FMT in the treatment of MDD. Numerous studies have shown a correlation between abnormalities of the gut microbiota and MDD, whereas FMT has demonstrated the potential to restore microbial equilibrium. While FMT has shown encouraging results, it is crucial to highlight the potential hazards and limits inherent to this therapeutic approach. Stool donor-to-recipient disease transfer is a concern of FMT. Furthermore, it still needs to be determined what effect FMT has on the gut microbiota and the brain in the long run. This literature review provides an overview of the possible efficacy of FMT as a therapeutic modality for MDD. There is hope for patients who have not reacted well to typical antidepressant therapy since FMT may become an invaluable tool in the treatment of MDD as researchers continue to examine the relationship between gut microbiota and MDD.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang 110042, China.
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang 110042, China
| |
Collapse
|
48
|
Fornari Laurindo L, Aparecido Dias J, Cressoni Araújo A, Torres Pomini K, Machado Galhardi C, Rucco Penteado Detregiachi C, Santos de Argollo Haber L, Donizeti Roque D, Dib Bechara M, Vialogo Marques de Castro M, de Souza Bastos Mazuqueli Pereira E, José Tofano R, Jasmin Santos German Borgo I, Maria Barbalho S. Immunological dimensions of neuroinflammation and microglial activation: exploring innovative immunomodulatory approaches to mitigate neuroinflammatory progression. Front Immunol 2024; 14:1305933. [PMID: 38259497 PMCID: PMC10800801 DOI: 10.3389/fimmu.2023.1305933] [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: 10/02/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
The increasing life expectancy has led to a higher incidence of age-related neurodegenerative conditions. Within this framework, neuroinflammation emerges as a significant contributing factor. It involves the activation of microglia and astrocytes, leading to the release of pro-inflammatory cytokines and chemokines and the infiltration of peripheral leukocytes into the central nervous system (CNS). These instances result in neuronal damage and neurodegeneration through activated nucleotide-binding domain and leucine-rich repeat containing (NLR) family pyrin domain containing protein 3 (NLRP3) and nuclear factor kappa B (NF-kB) pathways and decreased nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Due to limited effectiveness regarding the inhibition of neuroinflammatory targets using conventional drugs, there is challenging growth in the search for innovative therapies for alleviating neuroinflammation in CNS diseases or even before their onset. Our results indicate that interventions focusing on Interleukin-Driven Immunomodulation, Chemokine (CXC) Receptor Signaling and Expression, Cold Exposure, and Fibrin-Targeted strategies significantly promise to mitigate neuroinflammatory processes. These approaches demonstrate potential anti-neuroinflammatory effects, addressing conditions such as Multiple Sclerosis, Experimental autoimmune encephalomyelitis, Parkinson's Disease, and Alzheimer's Disease. While the findings are promising, immunomodulatory therapies often face limitations due to Immune-Related Adverse Events. Therefore, the conduction of randomized clinical trials in this matter is mandatory, and will pave the way for a promising future in the development of new medicines with specific therapeutic targets.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo, Brazil
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Jefferson Aparecido Dias
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Karina Torres Pomini
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Department of Anatomy, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Cristiano Machado Galhardi
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Claudia Rucco Penteado Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Luíza Santos de Argollo Haber
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Domingos Donizeti Roque
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Department of Anatomy, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Marcela Vialogo Marques de Castro
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Ricardo José Tofano
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
| | - Iris Jasmin Santos German Borgo
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, Universidade de São Paulo (FOB-USP), Bauru, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília, São Paulo, Brazil
| |
Collapse
|
49
|
Bou Sader Nehme S, Sanchez-Sarasua S, Adel R, Tuifua M, Ali A, Essawy AE, Abdel Salam S, Hleihel W, Boué-Grabot E, Landry M. P2X4 signalling contributes to hyperactivity but not pain sensitization comorbidity in a mouse model of attention deficit/hyperactivity disorder. Front Pharmacol 2024; 14:1288994. [PMID: 38239187 PMCID: PMC10794506 DOI: 10.3389/fphar.2023.1288994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction: Attention deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental disorder characterized by hyperactivity, inattention, and impulsivity that often persist until adulthood. Frequent comorbid disorders accompany ADHD and two thirds of children diagnosed with ADHD also suffer from behavioural disorders and from alteration of sensory processing. We recently characterized the comorbidity between ADHD-like symptoms and pain sensitisation in a pharmacological mouse model of ADHD, and we demonstrated the implication of the anterior cingulate cortex and posterior insula. However, few studies have explored the causal mechanisms underlying the interactions between ADHD and pain. The implication of inflammatory mechanisms has been suggested but the signalling pathways involved have not been explored. Methods: We investigated the roles of purinergic signalling, at the crossroad of pain and neuroinflammatory pathways, by using a transgenic mouse line that carries a total deletion of the P2X4 receptor. Results: We demonstrated that P2X4 deletion prevents hyperactivity in the mouse model of ADHD. In contrast, the absence of P2X4 lowered thermal pain thresholds in sham conditions and did not affect pain sensitization in ADHD-like conditions. We further analysed microglia reactivity and the expression of inflammatory markers in wild type and P2X4KO mice. Our results revealed that P2X4 deletion limits microglia reactivity but at the same time exerts proinflammatory effects in the anterior cingulate cortex and posterior insula. Conclusion: This dual role of P2X4 could be responsible for the differential effects noted on ADHD-like symptoms and pain sensitization and calls for further studies to investigate the therapeutic benefit of targeting the P2X4 receptor in ADHD patients.
Collapse
Affiliation(s)
- Sarah Bou Sader Nehme
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - Sandra Sanchez-Sarasua
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
- Faculty of Health Sciences, University of Jaume I, Castellon, Spain
| | - Ramy Adel
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marie Tuifua
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| | - Awatef Ali
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Amina E. Essawy
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Sherine Abdel Salam
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Walid Hleihel
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - Eric Boué-Grabot
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| | - Marc Landry
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| |
Collapse
|
50
|
Panbhare K, Pandey R, Chauhan C, Sinha A, Shukla R, Kaundal RK. Role of NLRP3 Inflammasome in Stroke Pathobiology: Current Therapeutic Avenues and Future Perspective. ACS Chem Neurosci 2024; 15:31-55. [PMID: 38118278 DOI: 10.1021/acschemneuro.3c00536] [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] [Indexed: 12/22/2023] Open
Abstract
Neuroinflammation is a key pathophysiological feature of stroke-associated brain injury. A local innate immune response triggers neuroinflammation following a stroke via activating inflammasomes. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has been heavily implicated in stroke pathobiology. Following a stroke, several stimuli have been suggested to trigger the assembly of the NLRP3 inflammasome. Recent studies have advanced the understanding and revealed several new players regulating NLRP3 inflammasome-mediated neuroinflammation. This article discussed recent advancements in NLRP3 assembly and highlighted stroke-induced mitochondrial dysfunction as a major checkpoint to regulating NLRP3 activation. The NLRP3 inflammasome activation leads to caspase-1-dependent maturation and release of IL-1β, IL-18, and gasdermin D. In addition, genetic or pharmacological inhibition of the NLRP3 inflammasome activation and downstream signaling has been shown to attenuate brain infarction and improve the neurological outcome in experimental models of stroke. Several drug-like small molecules targeting the NLRP3 inflammasome are in different phases of development as novel therapeutics for various inflammatory conditions, including stroke. Understanding how these molecules interfere with NLRP3 inflammasome assembly is paramount for their better optimization and/or development of newer NLRP3 inhibitors. In this review, we summarized the assembly of the NLRP3 inflammasome and discussed the recent advances in understanding the upstream regulators of NLRP3 inflammasome-mediated neuroinflammation following stroke. Additionally, we critically examined the role of the NLRP3 inflammasome-mediated signaling in stroke pathophysiology and the development of therapeutic modalities to target the NLRP3 inflammasome-related signaling for stroke treatment.
Collapse
Affiliation(s)
- Kartik Panbhare
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Rukmani Pandey
- Department of Psychiatry, Center for Molecular Biology and Genetics of Neurodegeneration, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Chandan Chauhan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Antarip Sinha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, UP 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| |
Collapse
|