1
|
Jha D, Bakker ENTP, Kumar R. Mechanistic and therapeutic role of NLRP3 inflammasome in the pathogenesis of Alzheimer's disease. J Neurochem 2024; 168:3574-3598. [PMID: 36802053 DOI: 10.1111/jnc.15788] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/20/2023]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, has emerged as the most common form of dementia in the elderly. Several pathological hallmarks have been identified, including neuroinflammation. A comprehensive insight into the underlying mechanisms that can fuel the development of novel therapeutic approaches is necessary because of the alarmingly rapid increase in the frequency of incidence. Recently, NLRP3 inflammasome was identified as a critical mediator of neuroinflammation. Activation of nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome by amyloid, neurofibrillary tangles, impaired autophagy and endoplasmic reticulum stress, triggers the release of pro-inflammatory cytokines such as IL-1β and IL-18. Subsequently, these cytokines can promote neurodegeneration and cognitive impairment. It is well established that genetic or pharmacological ablation of NLRP3 alleviates AD-related pathological features in in vitro and in vivo models. Therefore, several synthetic and natural compounds have been identified that exhibit the potential to inhibit NLRP3 inflammasome and alleviate AD-associated pathology. The current review article will highlight the various mechanisms by which activation of NLRP3 inflammation occurs during Alzheimer's disease, and how it influences neuroinflammation, neurodegeneration and cognitive impairment. Moreover, we will summarise the different small molecules that possess the potential to inhibit NLRP3 and can pave the path for developing novel therapeutic interventions for AD.
Collapse
Affiliation(s)
- Dhanshree Jha
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centre, Location University of Amsterdam, and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| | - Rahul Kumar
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centre, Location University of Amsterdam, and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Zhou X, Wang J, Yu L, Qiao G, Qin D, Yuen-Kwan Law B, Ren F, Wu J, Wu A. Mitophagy and cGAS-STING crosstalk in neuroinflammation. Acta Pharm Sin B 2024; 14:3327-3361. [PMID: 39220869 PMCID: PMC11365416 DOI: 10.1016/j.apsb.2024.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 09/04/2024] Open
Abstract
Mitophagy, essential for mitochondrial health, selectively degrades damaged mitochondria. It is intricately linked to the cGAS-STING pathway, which is crucial for innate immunity. This pathway responds to mitochondrial DNA and is associated with cellular stress response. Our review explores the molecular details and regulatory mechanisms of mitophagy and the cGAS-STING pathway. We critically evaluate the literature demonstrating how dysfunctional mitophagy leads to neuroinflammatory conditions, primarily through the accumulation of damaged mitochondria, which activates the cGAS-STING pathway. This activation prompts the production of pro-inflammatory cytokines, exacerbating neuroinflammation. This review emphasizes the interaction between mitophagy and the cGAS-STING pathways. Effective mitophagy may suppress the cGAS-STING pathway, offering protection against neuroinflammation. Conversely, impaired mitophagy may activate the cGAS-STING pathway, leading to chronic neuroinflammation. Additionally, we explored how this interaction influences neurodegenerative disorders, suggesting a common mechanism underlying these diseases. In conclusion, there is a need for additional targeted research to unravel the complexities of mitophagy-cGAS-STING interactions and their role in neurodegeneration. This review highlights potential therapies targeting these pathways, potentially leading to new treatments for neuroinflammatory and neurodegenerative conditions. This synthesis enhances our understanding of the cellular and molecular foundations of neuroinflammation and opens new therapeutic avenues for neurodegenerative disease research.
Collapse
Affiliation(s)
- Xiaogang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jing Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Gan Qiao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
3
|
Zhu T, Yao Y, Ding J, Zhang C, Xia N, Tao Y, Zhang W, Qi H, Gong L, Jiang P. 3-Methyladenine attenuates neuroinflammation and improves cognitive function in sepsis-associated encephalopathy by inhibiting autophagy. Int Immunopharmacol 2024; 139:112744. [PMID: 39059098 DOI: 10.1016/j.intimp.2024.112744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/01/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE Sepsis-associated encephalopathy (SAE) can lead to severe cerebral dysfunction as well as cognitive dysfunction, resulting in a significant disease burden. 3-Methyladenine (3-MA) has been confirmed to have anti-inflammatory effects on diseases characterized by enhanced autophagy. However, its role in SAE has not been clarified. METHODS An SAE mouse model was generated by intraperitoneal injection of lipopolysaccharide (LPS). Mice were given 5, 20, or 80 mg/kg 3-MA to determine the therapeutic dose. The mice in the different groups were given 20 mg/kg 3-MA or saline, and survival, body temperature, body weight and neurobehavioral scores were measured at different time points. The expression of autophagy-related proteins and inflammatory factors was detected by Western blotting, enzyme linked immunosorbent assay (ELISA) and real-time quantitative polymerase chain reaction (RT-qPCR) 12 h after LPS induction. Glial activation and neuronal injury in the hippocampus were detected by immunofluorescence staining and HE staining. The open Field test, novel object recognition (NOR) test, Y-maze test, and Morris water maze (MWM) test were performed to assess cognitive function. RESULTS Treatment with 20 or 80 mg/kg 3-MA reduced the increase in hippocampal TNF-α, IL-6, and IL-1β expression in SAE model mice, with 20 mg/kg 3-MA having the greatest therapeutic effect. Treatment with 20 mg/kg 3-MA effectively reduced the expression of hippocampal autophagy-related proteins and mortality, ameliorated hypothermia, decreased body weight and electroencephalography (EEG) performance, and attenuated the activation of neuroglia and neuronal damage. Moreover, it alleviated the cognitive dysfunction 2 weeks after LPS induction. CONCLUSIONS 3-MA reduced neuroglial activation and neuronal damage, attenuated neuroinflammation, and improved cognitive deficits during recovery period by inhibiting autophagy in SAE.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310020, China
| | - Yinping Yao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China; Department of Pediatrics, Shaoxing People's Hospital, Shaoxing, Zhejiang Province 312300, China
| | - Junchao Ding
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China; Department of Pediatrics, Yiwu Maternal and Child Health Care Hospital, Yiwu, Zhejiang Province 322000, China
| | - Chengyue Zhang
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China
| | - Ningxiao Xia
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China
| | - Yilin Tao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China
| | - Wenhao Zhang
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China
| | - Hantao Qi
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China
| | - Lifen Gong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China.
| | - Peifang Jiang
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province 310052, China.
| |
Collapse
|
4
|
Hadi F, Mortaja M, Hadi Z. Calcium (Ca 2+) hemostasis, mitochondria, autophagy, and mitophagy contribute to Alzheimer's disease as early moderators. Cell Biochem Funct 2024; 42:e4085. [PMID: 38951992 DOI: 10.1002/cbf.4085] [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: 11/04/2023] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
This review rigorously investigates the early cerebral changes associated with Alzheimer's disease, which manifest long before clinical symptoms arise. It presents evidence that the dysregulation of calcium (Ca2+) homeostasis, along with mitochondrial dysfunction and aberrant autophagic processes, may drive the disease's progression during its asymptomatic, preclinical stage. Understanding the intricate molecular interplay that unfolds during this critical period offers a window into identifying novel therapeutic targets, thereby advancing the treatment of neurodegenerative disorders. The review delves into both established and emerging insights into the molecular alterations precipitated by the disruption of Ca2+ balance, setting the stage for cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Fatemeh Hadi
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Mahsa Mortaja
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Zahra Hadi
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, Tehran, Iran
| |
Collapse
|
5
|
Yun Q, Ma SF, Zhang WN, Gu M, Wang J. FoxG1 as a Potential Therapeutic Target for Alzheimer's Disease: Modulating NLRP3 Inflammasome via AMPK/mTOR Autophagy Pathway. Cell Mol Neurobiol 2024; 44:35. [PMID: 38630150 PMCID: PMC11023968 DOI: 10.1007/s10571-024-01467-4] [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: 11/28/2023] [Accepted: 02/27/2024] [Indexed: 04/19/2024]
Abstract
An increasing body of research suggests that promoting microglial autophagy hinders the neuroinflammation initiated though the NLRP3 inflammasome activation in Alzheimer's disease (AD). The function of FoxG1, a crucial transcription factor involved in cell survival by regulating mitochondrial function, remains unknown during the AD process and neuroinflammation occurs. In the present study, we firstly found that Aβ peptides induced AD-like neuroinflammation upregulation and downregulated the level of autophagy. Following low-dose Aβ25-35 stimulation, FoxG1 expression and autophagy exhibited a gradual increase. Nevertheless, with high-concentration Aβ25-35 treatment, progressive decrease in FoxG1 expression and autophagy levels as the concentration of Aβ25-35 escalated. In addition, FoxG1 has a positive effect on cell viability and autophagy in the nervous system. In parallel with the Aβ25-35 stimulation, we employed siRNA to decrease the expression of FoxG1 in N2A cells. A substantial reduction in autophagy level (Beclin1, LC3II, SQSTM1/P62) and a notable growth in inflammatory response (NLRP3, TNF-α, and IL-6) were observed. In addition, we found FoxG1 overexpression owned the effect on the activation of AMPK/mTOR autophagy pathway and siRNA-FoxG1 successfully abolished this effect. Lastly, FoxG1 suppressed the NLRP3 inflammasome and enhanced the cognitive function in AD-like mouse model induced by Aβ25-35. Confirmed by cellular and animal experiments, FoxG1 suppressed NLRP3-mediated neuroinflammation, which was strongly linked to autophagy regulated by AMPK/mTOR. Taken together, FoxG1 may be a critical node in the pathologic progression of AD and has the potential to serve as therapeutic target.
Collapse
Affiliation(s)
- Qi Yun
- Changzhou Children's Hospital Affiliated to Nantong University, 958 Zhongwu Avenue, Changzhou, 213000, Jiangsu Province, China
| | - Si-Fei Ma
- Changzhou Blood Center, 118 Canal Road, Changzhou, 213000, Jiangsu Province, China
| | - Wei-Ning Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 213000, Jiangsu Province, China
| | - Meng Gu
- Changzhou Children's Hospital Affiliated to Nantong University, 958 Zhongwu Avenue, Changzhou, 213000, Jiangsu Province, China.
| | - Jia Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 213000, Jiangsu Province, China.
- The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu Province, PR China.
| |
Collapse
|
6
|
Khedr LH, Rahmo RM, Eldemerdash OM, Helmy EM, Ramzy FA, Lotfy GH, Zakaria HA, Gad MM, Youhanna MM, Samaan MH, Thabet NW, Ghazal RH, Rabie MA. Implication of M2 macrophage on NLRP3 inflammasome signaling in mediating the neuroprotective effect of Canagliflozin against methotrexate-induced cognitive impairment. Int Immunopharmacol 2024; 130:111709. [PMID: 38377857 DOI: 10.1016/j.intimp.2024.111709] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Methotrexate (MTX), a chemotherapeutic antimetabolite, has been linked to cognitive impairment in cancer patients. MTX-induced metabolic pathway disruption may result in decreased antioxidant activity and increased oxidative stress, influencing hippocampal neurogenesis and microglial activation. Nuclear factor-kappa B (NF-κB), an oxidative stress byproduct, has been linked to MTX toxicity via the activation of NLRP3 inflammasome signaling. Macrophage activation and polarization plays an important role in tissue injury. This differentiation may be mediated via either the Toll-like receptor 4 (TLR4) or NLRP3 inflammasome. Interestingly, Canagliflozin (CANA), a sodium-glucose cotransporter 2 (SGLT2) inhibitor has been recently reported to exert anti-inflammatory effects by modulating macrophage polarization balance. This study aimed to investigate CANA's protective effect against MTX-induced cognitive impairment, highlighting the possible involvement of TLR4/ NF-κB crosstalk with NLRP3 inflammasome activation and macrophage polarization. Forty-eight Male Wistar rats were divided into 4 groups; (1) received saline orally for 30 days and intravenously on days 8 and 15. (2) received Canagliflozin (CANA; 20 mg/kg/day; p.o.) for 30 days. (3) received MTX (75 mg/kg, i.v.) on day 8 and 15, then they were injected with four i.p. injections of leucovorin (LCV): the first dose was 6 mg/ kg after 18 h, and the remaining doses were 3 mg/kg after 26, 42, and 50 h of MTX administration. (4) received MTX and LCV as in group 3 in addition to CANA as in group 2. MTX-treated rats showed cognitive deficits in spatial and learning memory as evidenced in the novel object recognition and Morris water maze tests. MTX exerted an oxidative effect which was evident by the increase in MDA and decline in SOD, GSH and GPx. Moreover, it exerted an inflammatory effect via elevated caspase-1, IL-1β and IL-8. CANA treatment restored cognitive ability, reduced MTX-induced oxidative stress and neuroinflammation via attenuation of TLR4/NF-κB/NLRP3 signaling, and rebalanced macrophage polarization by promoting the M2 phenotype. Hence, targeting molecular mechanisms manipulating macrophage polarization may offer novel neuroprotective strategies for preventing or treating MTX-induced immune modulation and its detrimental sequel.
Collapse
Affiliation(s)
- Lobna H Khedr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Rania M Rahmo
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Omar M Eldemerdash
- Department of Biochemistry, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Engy M Helmy
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Felopateer A Ramzy
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - George H Lotfy
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Habiba A Zakaria
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Marine M Gad
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Marina M Youhanna
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Manar H Samaan
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Nevert W Thabet
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Reem H Ghazal
- Pharmacy Senior Students, Faculty of Pharmacy, Misr International University (MIU), Cairo 44971, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt.
| |
Collapse
|
7
|
Li Y, Xin Y, Qi MM, Wu ZY, Wang H, Zheng WC, Wang JX, Zhang DX, Zhang LM. VX-765 Alleviates Circadian Rhythm Disorder in a Rodent Model of Traumatic Brain Injury Plus Hemorrhagic Shock and Resuscitation. J Neuroimmune Pharmacol 2024; 19:3. [PMID: 38300393 DOI: 10.1007/s11481-024-10102-4] [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: 11/05/2021] [Accepted: 01/02/2024] [Indexed: 02/02/2024]
Abstract
Severe traumatic brain injury (TBI) can result in persistent complications, including circadian rhythm disorder, that substantially affect not only the injured people, but also the mood and social interactions with the family and the community. Pyroptosis in GFAP-positive astrocytes plays a vital role in inflammatory changes post-TBI. We determined whether VX-765, a low molecular weight caspase-1 inhibitor, has potential therapeutic value against astrocytic inflammation and pyroptosis in a rodent model of TBI plus hemorrhagic shock and resuscitation (HSR). A weight-drop plus bleeding and refusion model was used to establish traumatic exposure in rats. VX-765 (50 mg/kg) was injected via the femoral vein after resuscitation. Wheel-running activity was assessed, brain magnetic resonance images were evaluated, the expression of pyroptosis-associated molecules including cleaved caspase-1, gasdermin D (GSDMD), and interleukin-18 (IL-18) in astrocytes in the region of anterior hypothalamus, were explored 30 days post-trauma. VX-765-treated rats had significant improvement in circadian rhythm disorder, decreased mean diffusivity (MD) and mean kurtosis (MK), increased fractional anisotropy (FA), an elevated number and branches of astrocytes, and lower cleaved caspase-1, GSDMD, and IL-18 expression in astrocytes than TBI + HSR-treated rats. These results demonstrated that inhibition of pyroptosis-associated astrocytic activations in the anterior hypothalamus using VX-765 may ameliorate circadian rhythm disorder after trauma. In conclusion, we suggest that interventions targeting caspase-1-induced astrocytic pyroptosis by VX-765 are promising strategies to alleviate circadian rhythm disorder post-TBI.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Yue Xin
- Department of Anesthesiology, Graduated School, Hebei Medical University, Cangzhou, China
| | - Man-Man Qi
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Zhi-You Wu
- Department of Neurosurgery, Graduated School, Hebei Medical University, Cangzhou, China
| | - Han Wang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Wei-Chao Zheng
- Department of Anesthesiology, Graduated School, Hebei Medical University, Cangzhou, China
| | - Jie-Xia Wang
- Department of Anesthesiology, Graduated School, Hebei Medical University, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China.
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China.
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China.
| |
Collapse
|
8
|
Chu J, Zhang W, Liu Y, Gong B, Ji W, Yin T, Gao C, Liangwen D, Hao M, Chen C, Zhuang J, Gao J, Yin Y. Biomaterials-based anti-inflammatory treatment strategies for Alzheimer's disease. Neural Regen Res 2024; 19:100-115. [PMID: 37488851 PMCID: PMC10479833 DOI: 10.4103/1673-5374.374137] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/28/2023] [Accepted: 03/28/2023] [Indexed: 07/26/2023] Open
Abstract
The current therapeutic drugs for Alzheimer's disease only improve symptoms, they do not delay disease progression. Therefore, there is an urgent need for new effective drugs. The underlying pathogenic factors of Alzheimer's disease are not clear, but neuroinflammation can link various hypotheses of Alzheimer's disease; hence, targeting neuroinflammation may be a new hope for Alzheimer's disease treatment. Inhibiting inflammation can restore neuronal function, promote neuroregeneration, reduce the pathological burden of Alzheimer's disease, and improve or even reverse symptoms of Alzheimer's disease. This review focuses on the relationship between inflammation and various pathological hypotheses of Alzheimer's disease; reports the mechanisms and characteristics of small-molecule drugs (e.g., nonsteroidal anti-inflammatory drugs, neurosteroids, and plant extracts); macromolecule drugs (e.g., peptides, proteins, and gene therapeutics); and nanocarriers (e.g., lipid-based nanoparticles, polymeric nanoparticles, nanoemulsions, and inorganic nanoparticles) in the treatment of Alzheimer's disease. The review also makes recommendations for the prospective development of anti-inflammatory strategies based on nanocarriers for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jianjian Chu
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Weicong Zhang
- School of Pharmacy, University College London, London, UK
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baofeng Gong
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Chao Gao
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Danqi Liangwen
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Mengqi Hao
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Cuimin Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital (Shanghai Changzheng Hospital) of Naval Medical University, Shanghai, China
| |
Collapse
|
9
|
Hamzeh O, Rabiei F, Shakeri M, Parsian H, Saadat P, Rostami-Mansoor S. Mitochondrial dysfunction and inflammasome activation in neurodegenerative diseases: Mechanisms and therapeutic implications. Mitochondrion 2023; 73:S1567-7249(23)00087-9. [PMID: 39492438 DOI: 10.1016/j.mito.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/02/2023] [Accepted: 10/28/2023] [Indexed: 11/05/2024]
Abstract
Impaired mitochondrial function is crucial to the pathogenesis of several neurodegenerative diseases. It causes the release of mitochondrial DNA (mtDNA), mitochondrial reactive oxygen species (mtROS), ATP, and cardiolipin, which activate the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. NLRP3 inflammasome is an important innate immune system element contributing to neuroinflammation and neurodegeneration. Therefore, targeting the NLRP3 inflammasome has become an interesting therapeutic approach for treating neurodegenerative diseases. This review describes the role of mitochondrial abnormalities and over-activated inflammasomes in the progression of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple sclerosis (MS), Amyotrophic lateral sclerosis (ALS), and Friedrich ataxia (FRDA). We also discuss the therapeutic strategies focusing on signaling pathways associated with inflammasome activation, which potentially alleviate neurodegenerative symptoms and impede disease progression.
Collapse
Affiliation(s)
- Olia Hamzeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Rabiei
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mahdi Shakeri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Payam Saadat
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sahar Rostami-Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
10
|
Chiarini A, Gui L, Viviani C, Armato U, Dal Prà I. NLRP3 Inflammasome’s Activation in Acute and Chronic Brain Diseases—An Update on Pathogenetic Mechanisms and Therapeutic Perspectives with Respect to Other Inflammasomes. Biomedicines 2023; 11:biomedicines11040999. [PMID: 37189617 DOI: 10.3390/biomedicines11040999] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Increasingly prevalent acute and chronic human brain diseases are scourges for the elderly. Besides the lack of therapies, these ailments share a neuroinflammation that is triggered/sustained by different innate immunity-related protein oligomers called inflammasomes. Relevant neuroinflammation players such as microglia/monocytes typically exhibit a strong NLRP3 inflammasome activation. Hence the idea that NLRP3 suppression might solve neurodegenerative ailments. Here we review the recent Literature about this topic. First, we update conditions and mechanisms, including RNAs, extracellular vesicles/exosomes, endogenous compounds, and ethnic/pharmacological agents/extracts regulating NLRP3 function. Second, we pinpoint NLRP3-activating mechanisms and known NLRP3 inhibition effects in acute (ischemia, stroke, hemorrhage), chronic (Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, MS, ALS), and virus-induced (Zika, SARS-CoV-2, and others) human brain diseases. The available data show that (i) disease-specific divergent mechanisms activate the (mainly animal) brains NLRP3; (ii) no evidence proves that NLRP3 inhibition modifies human brain diseases (yet ad hoc trials are ongoing); and (iii) no findings exclude that concurrently activated other-than-NLRP3 inflammasomes might functionally replace the inhibited NLRP3. Finally, we highlight that among the causes of the persistent lack of therapies are the species difference problem in disease models and a preference for symptomatic over etiologic therapeutic approaches. Therefore, we posit that human neural cell-based disease models could drive etiological, pathogenetic, and therapeutic advances, including NLRP3’s and other inflammasomes’ regulation, while minimizing failure risks in candidate drug trials.
Collapse
|
11
|
Mishra P, Davies DA, Albensi BC. The Interaction Between NF-κB and Estrogen in Alzheimer's Disease. Mol Neurobiol 2023; 60:1515-1526. [PMID: 36512265 DOI: 10.1007/s12035-022-03152-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
Post-menopausal women are at a higher risk of developing Alzheimer's disease (AD) than males. The higher rates of AD in women are associated with the sharp decline in the estrogen levels after menopause. Estrogen has been shown to downregulate inflammatory cytokines in the central nervous system (CNS), which has a neuroprotective role against neurodegenerative diseases including AD. Sustained neuroinflammation is associated with neurodegeneration and contributes to AD. Nuclear factor kappa-B (NF-κB) is a transcription factor involved with the modulation of inflammation and interacts with estrogen to influence the progression of AD. Application of 17β-estradiol (E2) has been shown to inhibit NF-κB, thereby reducing transcription of NF-κB target genes. Despite accumulating evidence showing that estrogens have beneficial effects in pre-clinical AD studies, there are mixed results with hormone replacement therapy in clinical trials. Furthering our understanding of how NF-κB interacts with estrogen and alters the progression of neurodegenerative disorders including AD, should be beneficial and result in the development of novel therapeutics.
Collapse
Affiliation(s)
- Pranav Mishra
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada.,Department of Pharmacology & Therapeutics, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Don A Davies
- Department of Biology, York University, Toronto, ON, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada. .,Department of Pharmacology & Therapeutics, College of Medicine, University of Manitoba, Winnipeg, MB, Canada. .,Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.
| |
Collapse
|
12
|
Moonen S, Koper MJ, Van Schoor E, Schaeverbeke JM, Vandenberghe R, von Arnim CAF, Tousseyn T, De Strooper B, Thal DR. Pyroptosis in Alzheimer's disease: cell type-specific activation in microglia, astrocytes and neurons. Acta Neuropathol 2023; 145:175-195. [PMID: 36481964 DOI: 10.1007/s00401-022-02528-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
The major neuropathological hallmarks of Alzheimer's disease (AD) are amyloid β (Aβ) plaques and neurofibrillary tangles (NFT), accompanied by neuroinflammation and neuronal loss. Increasing evidence is emerging for the activation of the canonical NOD-, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome in AD. However, the mechanisms leading to neuronal loss in AD and the involvement of glial cells in these processes are still not clear. The aim of this study was to investigate the contribution of pyroptosis, a pro-inflammatory mechanism of cell death downstream of the inflammasome, to neurodegeneration in AD. Immunohistochemistry and biochemical analysis of protein levels were performed on human post-mortem brain tissue. We investigated the presence of cleaved gasdermin D (GSDMD), the pyroptosis effector protein, as well as the NLRP3 inflammasome-forming proteins, in the medial temporal lobe of 23 symptomatic AD, 25 pathologically defined preclinical AD (p-preAD) and 21 non-demented control cases. Cleaved GSDMD was detected in microglia, but also in astrocytes and in few pyramidal neurons in the first sector of the cornu ammonis (CA1) of the hippocampus and the temporal cortex of Brodmann area 36. Only microglia expressed all NLRP3 inflammasome-forming proteins (i.e., ASC, NLRP3, caspase-1). Cleaved GSDMD-positive astrocytes and neurons exhibited caspase-8 and non-canonical inflammasome protein caspase-4, respectively, potentially indicating alternative pathways for GSDMD cleavage. Brains of AD patients exhibited increased numbers of cleaved GSDMD-positive cells. Cleaved GSDMD-positive microglia and astrocytes were found in close proximity to Aβ plaques, while cleaved GSDMD-positive neurons were devoid of NFTs. In CA1, NLRP3-positive microglia and cleaved GSDMD-positive neurons were associated with local neuronal loss, indicating a possible contribution of NLRP3 inflammasome and pyroptosis activation to AD-related neurodegeneration. Taken together, our results suggest cell type-specific activation of pyroptosis in AD and extend the current knowledge about the contribution of neuroinflammation to the neurodegenerative process in AD via a direct link to neuron death by pyroptosis.
Collapse
Affiliation(s)
- Sebastiaan Moonen
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium.
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium.
| | - Marta J Koper
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Evelien Van Schoor
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium
- Laboratory for Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Jolien M Schaeverbeke
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Christine A F von Arnim
- Department of Neurology, University of Ulm, Ulm, Germany
- Department of Geriatrics, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Tousseyn
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium.
- Department of Pathology, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Guo B, Chen JH, Zhang JH, Fang Y, Liu XJ, Zhang J, Zhu HQ, Zhan L. Pattern-recognition receptors in endometriosis: A narrative review. Front Immunol 2023; 14:1161606. [PMID: 37033937 PMCID: PMC10076794 DOI: 10.3389/fimmu.2023.1161606] [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: 02/08/2023] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Endometriosis is closely associated with ectopic focal inflammation and immunosuppressive microenvironment. Multiple types of pattern recognition receptors (PRRs) are present in the innate immune system, which are able to detect pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) in both intracellular and external environments. However, the exact role of PRRs in endometriosis and the underlying molecular mechanism are unclear. PRRs are necessary for the innate immune system to identify and destroy invasive foreign infectious agents. Mammals mainly have two types of microbial recognition systems. The first one consists of the membrane-bound receptors, such as toll-like receptors (TLRs), which recognize extracellular microorganisms and activate intracellular signals to stimulate immune responses. The second one consists of the intracellular PRRs, including nod-like receptors (NLRs) and antiviral proteins retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA-5) with helix enzyme domain. In this review, we mainly focus on the key role of PRRs in the pathological processes associated with endometriosis. PRRs recognize PAMPs and can distinguish pathogenic microorganisms from self, triggering receptor ligand reaction followed by the stimulation of host immune response. Activated immune response promotes the transmission of microbial infection signals to the cells. As endometriosis is characterized by dysregulated inflammation and immune response, PRRs may potentially be involved in the activation of endometriosis-associated inflammation and immune disorders. Toll-like receptor 2 (TLR2), toll-like receptor 3 (TLR3), toll-like receptor 4 (TLR4), nod-like receptor family caspase activation and recruitment domain (CARD) domain containing 5 (NLRC5), nod-like receptor family pyrin domain containing 3 (NLRP3), and c-type lectin receptors (CLRs) play essential roles in endometriosis development by regulating immune and inflammatory responses. Absent in melanoma 2 (AIM2)-like receptors (ALRs) and retinoic acid-inducible gene I-like receptors (RLRs) may be involved in the activation of endometriosis-associated immune and inflammation disorders. PRRs, especially TLRs, may serve as potential therapeutic targets for alleviating pain in endometriosis patients. PRRs and their ligands interact with the innate immune system to enhance inflammation in the stromal cells during endometriosis. Thus, targeting PRRs and their new synthetic ligands may provide new therapeutic options for treating endometriosis.
Collapse
Affiliation(s)
- Bao Guo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jia hua Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jun hui Zhang
- First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuan Fang
- First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiao jing Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hai qing Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Zhan
- Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- *Correspondence: Lei Zhan,
| |
Collapse
|
14
|
Role of NLRP3 Inflammasome and Its Inhibitors as Emerging Therapeutic Drug Candidate for Alzheimer's Disease: a Review of Mechanism of Activation, Regulation, and Inhibition. Inflammation 2023; 46:56-87. [PMID: 36006570 PMCID: PMC9403980 DOI: 10.1007/s10753-022-01730-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders. The etiology and pathology of AD are complicated, variable, and yet to be completely discovered. However, the involvement of inflammasomes, particularly the NLRP3 inflammasome, has been emphasized recently. NLRP3 is a critical pattern recognition receptor involved in the expression of immune responses and has been found to play a significant role in the development of various immunological and neurological disorders such as multiple sclerosis, ulcerative colitis, gout, diabetes, and AD. It is a multimeric protein which releases various cytokines and causes caspase-1 activation through the process known as pyroptosis. Increased levels of cytokines (IL-1β and IL-18), caspase-1 activation, and neuropathogenic stimulus lead to the formation of proinflammatory microglial M1. Progressive researches have also shown that besides loss of neurons, the pathophysiology of AD primarily includes amyloid beta (Aβ) accumulation, generation of oxidative stress, and microglial damage leading to activation of NLRP3 inflammasome that eventually leads to neuroinflammation and dementia. It has been suggested in the literature that suppressing the activity of the NLRP3 inflammasome has substantial potential to prevent, manage, and treat Alzheimer's disease. The present review discusses the functional composition, various models, signaling molecules, pathways, and evidence of NLRP3 activation in AD. The manuscript also discusses the synthetic drugs, their clinical status, and projected natural products as a potential therapeutic approach to manage and treat NLRP3 mediated AD.
Collapse
|
15
|
Zheng X, Gong T, Tang C, Zhong Y, Shi L, Fang X, Chen D, Zhu Z. Gastrodin improves neuroinflammation-induced cognitive dysfunction in rats by regulating NLRP3 inflammasome. BMC Anesthesiol 2022; 22:371. [PMID: 36456961 PMCID: PMC9714247 DOI: 10.1186/s12871-022-01915-y] [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: 08/03/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Neuroinflammation is the main pathological mechanism of cognitive dysfunction caused by neurodegenerative diseases, and effective preventive and therapeutic measures are not available. We predicted the key targets of gastrodin's effects upon neuroinflammation through Network Pharmacology and molecular docking. Then the predicted targets were used to study how gastrodin affected cognitive dysfunction triggered by lipopolysaccharide-induced neuroinflammation in rats and its mechanisms. Three-month-old male rats were intraperitoneally injected with lipopolysaccharide for 3 days (d), 7 d and 14 d respectively. Gastrodin improved learning and memory ability of rats with neuroinflammation. Lipopolysaccharide enhanced the levels of pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-6, in rat hippocampus, which could be reversed by gastrodin. Gastrodin also inhibited the activation of microglia. Our findings suggested that gastrodin exerted neuroprotective effects in rats with neuroinflammation by impacting the TLR4-NF-kB-NLRP3 pathway. Therefore, gastrodin may be a potential therapeutic agent for neuroinflammation-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Xue Zheng
- grid.263761.70000 0001 0198 0694Suzhou Medical College of Soochow University, Suzhou, 215000 Jiangsu Province China ,Department of Anesthesiology, Zunyi Maternal And Child Health Care Hospital, 287#, Zhonghua Road, Zunyi, 563000 Guizhou Province China
| | - Taowu Gong
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| | - Chunchun Tang
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| | - Yuanping Zhong
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| | - Lu Shi
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| | - Xu Fang
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| | - Dongqin Chen
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| | - Zhaoqiong Zhu
- grid.413390.c0000 0004 1757 6938Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149#, Dalian Road, Zunyi, 563000 Guizhou Province China
| |
Collapse
|
16
|
Proaño B, Casani-Cubel J, Benlloch M, Rodriguez-Mateos A, Navarro-Illana E, Lajara-Romance JM, de la Rubia Ortí JE. Is Dutasteride a Therapeutic Alternative for Amyotrophic Lateral Sclerosis? Biomedicines 2022; 10:biomedicines10092084. [PMID: 36140184 PMCID: PMC9495995 DOI: 10.3390/biomedicines10092084] [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: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that is characterized by the loss of upper and lower motor neurons (MNs) in the cerebral cortex, brainstem and spinal cord, with consequent weakness, atrophy and the progressive paralysis of all muscles. There is currently no medical cure, and riluzole and edaravone are the only two known approved drugs for treating this condition. However, they have limited efficacy, and hence there is a need to find new molecules. Dutasteride, a dual inhibitor of type 1 and type 2 5α-reductase (5AR) enzymes, the therapeutic purposes of which, to date, are the treatment of benign prostatic hyperplasia and androgenic alopecia, shows great anti-ALS properties by the molecular-topology methodology. Based on this evidence, this review aims to assess the effects of dutasteride on testosterone (T), progesterone (PROG) and 17β-estradiol (17BE) as a therapeutic alternative for the clinical improvement of ALS, based on the hormonal, metabolic and molecular pathways related to the pathogenesis of the disease. According to the evidence found, dutasteride shows great neuroprotective, antioxidant and anti-inflammatory effects. It also appears effective against glutamate toxicity, and it is capable of restoring altered dopamine activity (DA). These effects are achieved both directly and through steroid hormones. Therefore, dutasteride seems to be a promising molecule for the treatment of ALS, although clinical studies are required for confirmation.
Collapse
Affiliation(s)
- Belén Proaño
- Doctoral Degree School, Health Sciences, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
| | - Julia Casani-Cubel
- School of Medicine and Health Sciences, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - María Benlloch
- Department Nursing, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, UK
| | | | | | | |
Collapse
|
17
|
Mata-Martínez E, Díaz-Muñoz M, Vázquez-Cuevas FG. Glial Cells and Brain Diseases: Inflammasomes as Relevant Pathological Entities. Front Cell Neurosci 2022; 16:929529. [PMID: 35783102 PMCID: PMC9243488 DOI: 10.3389/fncel.2022.929529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/27/2022] [Indexed: 12/13/2022] Open
Abstract
Inflammation mediated by the innate immune system is a physiopathological response to diverse detrimental circumstances such as microbe infections or tissular damage. The molecular events that underlie this response involve the assembly of multiprotein complexes known as inflammasomes. These assemblages are essentially formed by a stressor-sensing protein, an adapter protein and a non-apoptotic caspase (1 or 11). The coordinated aggregation of these components mediates the processing and release of pro-inflammatory interleukins (IL-β and IL-18) and cellular death by pyroptosis induction. The inflammatory response is essential for the defense of the organism; for example, it triggers tissue repair and the destruction of pathogen microbe infections. However, when inflammation is activated chronically, it promotes diverse pathologies in the lung, liver, brain and other organs. The nervous system is one of the main tissues where the inflammatory process has been characterized, and its implications in health and disease are starting to be understood. Thus, the regulation of inflammasomes in specific cellular types of the central nervous system needs to be thoroughly understood to innovate treatments for diverse pathologies. In this review, the presence and participation of inflammasomes in pathological conditions in different types of glial cells will be discussed.
Collapse
|
18
|
P2X4 Inhibition reduces microglia inflammation and apoptosis by NLRP3 and improves nervous system defects in rat brain trauma model. J Clin Neurosci 2022; 99:224-232. [DOI: 10.1016/j.jocn.2022.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 12/14/2022]
|
19
|
Lee HJ, Park JH, Hoe HS. Idebenone Regulates Aβ and LPS-Induced Neurogliosis and Cognitive Function Through Inhibition of NLRP3 Inflammasome/IL-1β Axis Activation. Front Immunol 2022; 13:749336. [PMID: 35222363 PMCID: PMC8866241 DOI: 10.3389/fimmu.2022.749336] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
Idebenone is an analogue of coenzyme Q10, an electron donor in the mitochondrial electron transport chain, and thus may function as an antioxidant to facilitate mitochondrial function. However, whether idebenone modulates LPS- and Aβ-mediated neuroinflammatory responses and cognitive function in vivo is unknown. The present study explored the effects of idebenone on LPS- or Aβ-mediated neuroinflammation, learning and memory and the underlying molecular mechanisms in wild-type (WT) mice and 5xFAD mice, a mouse model of Alzheimer’s disease (AD). In male and female WT mice, idebenone upregulated neuroprotective NRF2 expression, rescued LPS-induced spatial and recognition memory impairments, and reduced NLRP3 priming and subsequent neuroinflammation. Moreover, idebenone downregulated LPS-mediated neurogliosis, reactive oxygen species (ROS) levels, and mitochondrial function in BV2 microglial cells and primary astrocytes by inhibiting NLRP3 inflammasome activation. In 5xFAD mice, idebenone increased neuroprotective NRF2 expression and improved amyloid beta (Aβ)-induced cognitive dysfunction. Idebenone downregulated Aβ-mediated gliosis and proinflammatory cytokine levels in 5xFAD mice by modulating the vicious NLRP3/caspase-1/IL-1β neuroinflammation cycle. Taken together, our results suggest that idebenone targets neuroglial NLRP3 inflammasome activation and therefore may have neuroprotective effects and inhibit the pathological progression of neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Jin-Hee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| |
Collapse
|
20
|
El-Maadawy WH, Hassan M, Abdou RM, El-Dine RS, Aboushousha T, El-Tanbouly ND, El-Sayed AM. 6-Paradol alleviates Diclofenac-induced acute kidney injury via autophagy enhancement-mediated by AMPK/AKT/mTOR and NLRP3 inflammasome pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 91:103817. [PMID: 35091105 DOI: 10.1016/j.etap.2022.103817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/10/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Diclofenac (DIC)-induced acute kidney injury (AKI) causes high morbidity and mortality. With the absence of satisfactory treatment, we investigated the protective effects of 6-Paradol (PDL) against DIC-induced AKI, with focus on renal autophagy and NLRP3 inflammasome pathways . PDL has anti-inflammatory, antioxidant and AMPK-activation properties. PDL was administered to DIC-challenged rats. Nephrotoxicity, oxidative stress, inflammatory, and autophagy markers and histopathological examinations were evaluated. Compared to DIC, PDL restored serum nephrotoxicity, renal oxidative stress and pro-inflammatory markers. PDL almost restored renal architecture, upregulated renal Nrf2 pathway via enhancing Nrf2 mRNA expression and HO-1 levels. PDL suppressed renal NF-κB mRNA expression, and NLRP3 inflammasome pathway expression. Moreover, PDL enhanced renal autophagy through upregulating LC3B, AMPK and SIRT-1, and suppressed mTOR, p-AKT mRNA expressions and phosphorylated-p62 levels. Our study confirmed that autophagy suppression mediates DIC-induced AKI via AMPK/mTOR/AKT and NLRP3-inflammasome pathways. Also, PDL's nephroprotective effects could provide a promising therapeutic approach against DIC-induced AKI.
Collapse
Affiliation(s)
- Walaa H El-Maadawy
- Department of Pharmacology, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt.
| | - Marwa Hassan
- Department of Immunology, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt
| | - Rabab M Abdou
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Riham S El-Dine
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| | - Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba (P.O. 30), Giza 12411, Egypt
| | - Nebal D El-Tanbouly
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| | - Aly M El-Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| |
Collapse
|
21
|
Liu C, Gao W, Zhao L, Cao Y. Progesterone attenuates neurological deficits and exerts a protective effect on damaged axons via the PI3K/AKT/mTOR-dependent pathway in a mouse model of intracerebral hemorrhage. Aging (Albany NY) 2022; 14:2574-2589. [PMID: 35305084 PMCID: PMC9004566 DOI: 10.18632/aging.203954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating event with high disability and fatality rates. However, there is a lack of effective treatments for this condition. We aimed to investigate the neuroprotective and axonal regenerative effects of progesterone after ICH. For this purpose, an ICH model was established in adult mice by injecting type VII collagenase into the striatum; the mice were then treated with progesterone (8 mg/kg). Hematoma absorption, neurological scores, and brain water content were evaluated on days one, three, and seven after the ICH. The effect of progesterone on inflammation and axonal regeneration was examined on day three after the ICH using western blotting, immunohistochemistry, immunofluorescence, as well as hematoxylin-eosin, Nissl, and Luxol fast blue staining. In addition, we combined progesterone with the phosphoinositide 3-kinase/serine/threonine-specific protein kinase (PI3K/AKT) inhibitor, LY294002, to explore its potential neuroprotective mechanisms. Administration of progesterone attenuated the neurological deficits and expression of inflammatory cytokines and promoted axonal regeneration after ICH, this effect was blocked by LY294002. Collectively, these results suggest that progesterone could reduce axonal damage and produced partial neuroprotective effects after ICH through the PI3K/AKT/mTOR pathway, providing a new therapeutic target and basis for the treatment of ICH.
Collapse
Affiliation(s)
- Chang Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, P.R. China
| | - Weina Gao
- Department of Intensive Care Unit, The Affiliated Chengdu 363 Hospital of Southwest Medical University, Chengdu 610041, Sichuan Province, P.R. China
| | - Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637002, Sichuan Province, P.R. China
| | - Yi Cao
- Department of Neurosurgery, Chengdu Second People's Hospital, Chengdu 610021, Sichuan Province, P.R. China
| |
Collapse
|
22
|
Liang T, Zhang Y, Wu S, Chen Q, Wang L. The Role of NLRP3 Inflammasome in Alzheimer’s Disease and Potential Therapeutic Targets. Front Pharmacol 2022; 13:845185. [PMID: 35250595 PMCID: PMC8889079 DOI: 10.3389/fphar.2022.845185] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/24/2022] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a common age-related neurodegenerative disease characterized by progressive cognitive dysfunction and behavioral impairment. The typical pathological characteristics of AD are extracellular senile plaques composed of amyloid ß (Aβ) protein, intracellular neurofibrillary tangles formed by the hyperphosphorylation of the microtubule-associated protein tau, and neuron loss. In the past hundred years, although human beings have invested a lot of manpower, material and financial resources, there is no widely recognized drug for the effective prevention and clinical cure of AD in the world so far. Therefore, evaluating and exploring new drug targets for AD treatment is an important topic. At present, researchers have not stopped exploring the pathogenesis of AD, and the views on the pathogenic factors of AD are constantly changing. Multiple evidence have confirmed that chronic neuroinflammation plays a crucial role in the pathogenesis of AD. In the field of neuroinflammation, the nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome is a key molecular link in the AD neuroinflammatory pathway. Under the stimulation of Aβ oligomers and tau aggregates, it can lead to the assembly and activation of NLRP3 inflammasome in microglia and astrocytes in the brain, thereby causing caspase-1 activation and the secretion of IL-1β and IL-18, which ultimately triggers the pathophysiological changes and cognitive decline of AD. In this review, we summarize current literatures on the activation of NLRP3 inflammasome and activation-related regulation mechanisms, and discuss its possible roles in the pathogenesis of AD. Moreover, focusing on the NLRP3 inflammasome and combining with the upstream and downstream signaling pathway-related molecules of NLRP3 inflammasome as targets, we review the pharmacologically related targets and various methods to alleviate neuroinflammation by regulating the activation of NLRP3 inflammasome, which provides new ideas for the treatment of AD.
Collapse
Affiliation(s)
- Tao Liang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suyuan Wu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Lin Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Lin Wang,
| |
Collapse
|
23
|
Yue Q, Zhou X, Zhang Z, Hoi MPM. Murine Beta-Amyloid (1-42) Oligomers Disrupt Endothelial Barrier Integrity and VEGFR Signaling via Activating Astrocytes to Release Deleterious Soluble Factors. Int J Mol Sci 2022; 23:ijms23031878. [PMID: 35163801 PMCID: PMC8836933 DOI: 10.3390/ijms23031878] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
Transgenic mouse models of Alzheimer’s disease (AD) overexpress mutations of the human amyloid protein precursor (APP) and presenilin-1 (PSEN1) genes, which are known causes of amyloid pathology in familial AD. However, animal models for studying AD in the context of aging and age-related co-morbidities, such as blood–brain barrier (BBB) disruptions, are lacking. More recently, aged and progeroid mouse models have been proposed as alternatives to study aging-related AD, but the toxicity of murine amyloid-beta protein (Aβ) is not well defined. In this study, we aimed to study the potential toxicity of murine Aβ on brain endothelial cells and astrocytes, which are important components of the BBB, using mouse brain endothelial cells (bEnd.3) and astrocytes (C8-D1A). Murine-soluble Aβ (1–42) oligomers (sAβO42) (10 µM) induced negligible injuries in an endothelial monolayer but induced significant barrier disruptions in a bEnd.3 and C8-D1A co-culture. Similar results of endothelial perturbation were observed in a bEnd.3 monolayer treated with astrocyte-conditioned medium (ACM) generated by astrocytes exposed to sAβO42 (ACM-sAβO42), while additional exogenous sAβO42 did not cause further damage. Western blot analysis showed that ACM-sAβO42 altered the basal activities of vascular endothelial growth factor receptor 2 (VEGFR2), eNOS, and the signaling of the MEK/ERK and Akt pathways in bEnd.3. Our results showed that murine sAβO42 was moderately toxic to an endothelial and astrocyte co-culture. These damaging effects on the endothelial barrier were induced by deleterious soluble factors released from astrocytes, which disrupted endothelial VEGFR2 signaling and perturbed cell survival and barrier stabilization.
Collapse
Affiliation(s)
- Qian Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China; (Q.Y.); (X.Z.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR 999078, China
| | - Xinhua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China; (Q.Y.); (X.Z.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR 999078, China
| | - Zaijun Zhang
- Institute of New Drug Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine & New Drug Research, College of Pharmacy, Jinan University, Guangzhou 510632, China;
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China; (Q.Y.); (X.Z.)
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR 999078, China
- Correspondence: ; Tel.: +853-8822-4876
| |
Collapse
|
24
|
Deng Z, Dong Y, Zhou X, Lu JH, Yue Z. Pharmacological modulation of autophagy for Alzheimer’s disease therapy: Opportunities and obstacles. Acta Pharm Sin B 2021; 12:1688-1706. [PMID: 35847516 PMCID: PMC9279633 DOI: 10.1016/j.apsb.2021.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent and deleterious neurodegenerative disorder characterized by an irreversible and progressive impairment of cognitive abilities as well as the formation of amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. By far, the precise mechanisms of AD are not fully understood and no interventions are available to effectively slow down progression of the disease. Autophagy is a conserved degradation pathway that is crucial to maintain cellular homeostasis by targeting damaged organelles, pathogens, and disease-prone protein aggregates to lysosome for degradation. Emerging evidence suggests dysfunctional autophagy clearance pathway as a potential cellular mechanism underlying the pathogenesis of AD in affected neurons. Here we summarize the current evidence for autophagy dysfunction in the pathophysiology of AD and discuss the role of autophagy in the regulation of AD-related protein degradation and neuroinflammation in neurons and glial cells. Finally, we review the autophagy modulators reported in the treatment of AD models and discuss the obstacles and opportunities for potential clinical application of the novel autophagy activators for AD therapy.
Collapse
Affiliation(s)
- Zhiqiang Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Yu Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Xiaoting Zhou
- Department of Neurology, the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
- Corresponding authors.
| | - Zhenyu Yue
- Department of Neurology, the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Corresponding authors.
| |
Collapse
|
25
|
Shi Q, Zheng YY, Wang L, Xue YD, Yang YL. Curcumin suppresses neuroinflammation to protect neurons by preventing NLRP3 inflammasome activation. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211058615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction Nucleotide-binding and oligomerization domain like receptors protein 3 (NLRP3) inflammasome-mediated interleukin (IL)-1β secretion plays an important role in the progression of Alzheimer’s disease (AD). Curcumin has been shown to improve cognitive impairment and learning ability of AD mice by reducing IL-1β secretion. However, its exact mechanism of action remains unclear. In the present study, we explored the relationship between the neuroprotective effect of curcumin and activation of the NLRP3 inflammasome pathway. Methods BV2 cells were primed with 500 ng/mL lipopolysaccharide (LPS) for 4 h and subsequently treated with 50 μM Aβ25-35 for 24 h or pretreated with 2.5–10 μM curcumin for 4 h and exposed to 50 μM Aβ25-35 for 24 h. The effects of curcumin and Aβ25-35 were assessed by the CCK8 assay. ELISA was used for the detection of IL-1β, IL-6, and tumor necrosis factor (TNF)-α levels in the supernatant of the cell culture medium. The viability of SH-SY5Y cells, which were incubated with conditioned medium (CM) was assessed using the CCK8 assay. The percentage of apoptotic SH-SY5Y cells incubated with CM was assessed using Annexin V-FITC/PI staining flow cytometry analysis. The expression levels of NLRP3, caspase-1 and IL-1β were observed by western blot and immunofluorescence staining analyses; the mRNA levels of nlrp3, caspase-1 and IL-1β were analyzed using qRT-PCR. Results Low (2.5 μM), medium (5 μM), and high (10 μM) concentrations of curcumin and 50 μM Aβ25-35 were used to perform the experiments in the present study. Curcumin attenuated the IL-1β, IL-6, and TNF-α release and increased SH-SY5Y cell activity, while decreasing the apoptotic percentage of SH-SY5Y cells using Aβ25-35 for cell stimulation ( p < 0.05). Furthermore, curcumin inhibited the expression of NLRP3, caspase-1 and IL-1β and nlrp3 in BV-2 cells ( p < 0.05), However, curcumin did not affect the expression levels of caspase-1 and IL-1β ( p > 0.05) Conclusion Overall, the data indicated that curcumin is a promising neuroprotective agent for suppressing neuroinflammation by inhibiting the NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Qiang Shi
- Yan'an University Affiliated Hospital, Yan'an, China
| | | | - Le Wang
- Yan'an University Affiliated Hospital, Yan'an, China
| | - Yi-dong Xue
- Yan'an University Affiliated Hospital, Yan'an, China
| | | |
Collapse
|
26
|
NLRP3 Inflammasome Is Involved in Cocaine-Mediated Potentiation on Behavioral Changes in CX3CR1-Deficient Mice. J Pers Med 2021; 11:jpm11100963. [PMID: 34683104 PMCID: PMC8540128 DOI: 10.3390/jpm11100963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/25/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022] Open
Abstract
Microglia, the primary immunocompetent cells of the brain, are suggested to play a role in the development of drug addiction. Previous studies have identified the microglia-derived pro-inflammatory factor IL1β can promote the progression of cocaine addiction. Additionally, the activation status of microglia and “two-hit hypothesis” have been proposed in the field of drug addiction to explain how early life stress (ELS) could significantly increase the incidence of drug addiction in later life. However, the mechanisms underlying microglia prime and full activation and their roles in drug addiction remain greatly unexplored. Here, we employed CX3CR1-GFP mice (CX3CR1 functional deficiency, CX3CR1−/−) to explore whether primed microglia could potentiate cocaine-mediated behavioral changes and the possible underlying mechanisms. CX3CR1−/− mice revealed higher hyperlocomotion activity and conditional place preference than wild-type (WT) mice did under cocaine administration. In parallel, CX3CR1−/− mice showed higher activity of NLR family pyrin domain-containing 3 (NLRP3) inflammasome than WT mice. Interestingly, CX3CR1 deficiency itself could prime NLRP3 signaling by increasing the expression of NLPR3 and affect lysosome biogenesis under basal conditions. Taken together, our findings demonstrated that the functional status of microglia could have an impact on cocaine-mediated reward effects, and NLRP3 inflammasome activity was associated with this phenomenon. This study was consistent with the two-hit hypothesis and provided solid evidence to support the involvement of microglia in drug addiction. Targeting the NLRP3 inflammasome may represent a novel therapeutic approach for ameliorating or blocking the development of drug addiction.
Collapse
|
27
|
Ashrafizadeh M, Najafi M, Kavyiani N, Mohammadinejad R, Farkhondeh T, Samarghandian S. Anti-Inflammatory Activity of Melatonin: a Focus on the Role of NLRP3 Inflammasome. Inflammation 2021; 44:1207-1222. [PMID: 33651308 DOI: 10.1007/s10753-021-01428-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/19/2022]
Abstract
Melatonin is a hormone of the pineal gland that contributes to the regulation of physiological activities, such as sleep, circadian rhythm, and neuroendocrine processes. Melatonin is found in several plants and has pharmacological activities including antioxidant, anti-inflammatory, hepatoprotective, cardioprotective, and neuroprotective. It also has shown therapeutic efficacy in treatment of cancer and diabetes. Melatonin affects several molecular pathways to exert its protective effects. The NLRP3 inflammasome is considered a novel target of melatonin. This inflammasome contributes to enhanced level of IL-1β, caspase-1 activation, and pyroptosis stimulation. The function of NLRP3 inflammasome has been explored in various diseases, including cancer, diabetes, and neurological disorders. By inhibiting NLRP3, melatonin diminishes inflammation and influences various molecular pathways, such as SIRT1, microRNA, long non-coding RNA, and Wnt/β-catenin. Here, we discuss these molecular pathways and suggest that melatonin-induced inhibition of NLRP3 should be advanced in disease therapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasim Kavyiani
- Department of Basic Science, Faculty of Veterinary Medicine Faculty, Islamic Azad Branch, University of Shushtar, Shushtar, Khuzestan, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
28
|
Bai H, Zhang Q. Activation of NLRP3 Inflammasome and Onset of Alzheimer's Disease. Front Immunol 2021; 12:701282. [PMID: 34381452 PMCID: PMC8350495 DOI: 10.3389/fimmu.2021.701282] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
The nucleotide-binding domain leucine-rich repeat and pyrin domain containing receptor protein 3 (NLRP3) is an important pattern recognition receptor in human innate immunity. Activation of the NLRP3 inflammasome play a key role in the pathogenesis of Alzheimer’s disease (AD). Theories explaining activation of the NLRP3 inflammasome include the reactive oxygen species theory, the lysosomal damage theory and the mitochondrial DNA theory. The NLRP3 activation promotes occurrence of AD by producing IL-1β, IL-18 and other cytokines, and then by affecting the deposition of Aβ and tau proteins. Over-activated NLRP3 inflammasome often impair cell function and induces immune-related diseases. Some mechanisms have been found to negatively regulate activation of the NLRP3 inflammasome, which may be through receptor binding blocking mechanism, autophagy related mechanism, abnormal cytokine secretion mechanism, or interference related gene expression regulation mechanism. In this review, we summarize the possible mechanisms by which the activation of NLRP3 inflammasomes affects the pathogenesis of AD, and the recent advances in the prevention and treatment of AD by controlling the activation of NLRP3 inflammasomes. By researching the activation or inactivation of NLRP3 inflammasome, it is possible to reveal the pathogenesis of AD from a new perspective and provide a new idea for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Hua Bai
- Department of Neurology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, China.,Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Medical Experimental Center of the Third Affiliated Hospital of Guizhou Medical University, Duyun, China
| | - Qifang Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China.,Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
29
|
Gastrodin alleviates inflammatory injury of cardiomyocytes in septic shock mice via inhibiting NLRP3 expression. In Vitro Cell Dev Biol Anim 2021; 57:571-581. [PMID: 34106415 DOI: 10.1007/s11626-021-00593-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 05/13/2021] [Indexed: 12/25/2022]
Abstract
Septic shock leads to myocardial dysfunction and induces inflammation. Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes are involved in inflammation, and gastrodin can inhibit the activity of inflammasomes. Our study aimed to explore the effect of gastrodin against septic shock-induced injury through inhibiting NLRP3. Before establishing septic shock mice model, the mice were injected with gastrodin of various concentrations. The cardiac function of mice was detected by a PowerLab, and the histopathological changes of mouse myocardial tissues were detected by hematoxylin-eosin staining. Apoptosis of cardiomyocytes from mice was detected by TUNEL assay, and IL-1β concentration was detected by enzyme-linked immunosorbent assay. After culture in vitro and treatment with gastrodin, lipopolysaccharide (LPS), and NLRP3 vector, the cell viability and apoptosis of cardiomyocytes were detected by cell counting kit-8 and flow cytometry respectively. Besides, the expressions of NLRP3, Caspase-1, IL-1β, Bax, and Bcl-2 in mouse myocardial tissue or cultured cardiomyocytes were detected by Western blot. Gastrodin improved survival and promoted the recovery of cardiac function in septic shock mice, as it reversed the abnormality of left ventricular function indices in septic shock mice. Besides, gastrodin decreased IL-1β concentration and apoptosis in myocardial tissues of septic shock mice and decreased apoptosis and increased cell viability in LPS-induced cardiomyocytes. In addition, gastrodin downregulated NLRP3, Caspase-1, IL-1β, and Bax expressions and upregulated Bcl-2 expression in myocardial tissues of septic shock mice and LPS-induced cardiomyocytes. NLRP3 overexpression reversed the effect of gastrodin on LPS-induced cardiomyocytes. Gastrodin promoted cardiac function recovery and protected cardiomyocytes against septic shock-induced injury by regulating NLRP3.
Collapse
|
30
|
Shi W, Wu H, Liu S, Wu Z, Wu H, Liu J, Hou Y. Progesterone Suppresses Cholesterol Esterification in APP/PS1 mice and a cell model of Alzheimer's Disease. Brain Res Bull 2021; 173:162-173. [PMID: 34044033 DOI: 10.1016/j.brainresbull.2021.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/27/2021] [Accepted: 05/21/2021] [Indexed: 01/01/2023]
Abstract
AIMS Cholesteryl ester(CE), generated from the mitochondria associated membrane (MAM), is involved in the pathogenesis of Alzheimer's Disease (AD). In theory, the different neuroprotective effects of progesterone in AD are all linked to MAM, yet the effect on cholesterol esterification has not been reported. Therefore, this study was aimed to investigate the regulation of progesterone on intracerebral CE in AD models and the underlying mechanism. METHODS APP/PS1 mice and AD cell model induced by Aβ 25-35 were selected as the research objects. APP/PS1 mice were daily administrated intragastrically with progesterone and The Morris Water Maze test was performed to detect the learning and memory abilities. Intracellular cholesterol was measured by Cholesterol/Cholesteryl Ester Quantitation Assay. The structure of MAMs were observed with transmission electron microscopy. The expression of acyl-CoA: cholesterol acyltransferase 1 (ACAT1), ERK1/2 and p-ERK1/2 were detected with western blotting, immunohistochemistry or immunofluorescence. RESULTS Progesterone suppressed the accumulation of intracellular CE, shortened the length of abnormally prolonged MAM in cortex of APP/PS1 mice. Progesterone decreased the expression of ACAT1, which could be blocked by progesterone receptor membrane component 1 (PGRMC1) inhibitor AG205. The ERK1/2 pathway maybe involved in the progesterone mediated regulation of ACAT1 in AD models, rather than the PI3K/Akt and the P38 MEPK pathways. SIGNIFICANCE The results supported a line of evidence that progesterone regulates CE level and the structure of MAM in neurons of AD models, providing a promising treatment against AD on the dysfunction of cholesterol metabolism.
Collapse
Affiliation(s)
- Wenjing Shi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Pharmacy, Hebei General Hospital, Shijiazhuang 050051, Hebei Province, China.
| | - Hang Wu
- Department of Pharmacy, Heze University, Heze 274000, Shandong Province, China.
| | - Sha Liu
- Department of Pharmacy, the Third Hospital of Shijiazhuang, Shijiazhuang 050000, Hebei Province, China.
| | - Zhigang Wu
- Department of Pharmacy, Hebei North University, Hebei Key Laboratory of Neuropharmacology, Zhangjiakou 075000, China.
| | - Honghai Wu
- Department of Pharmacy, Bethune International Peace Hospital, Shijiazhuang 050082, Hebei Province, China.
| | - Jianfang Liu
- Department of Pharmacy, Bethune International Peace Hospital, Shijiazhuang 050082, Hebei Province, China.
| | - Yanning Hou
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Pharmacy, Bethune International Peace Hospital, Shijiazhuang 050082, Hebei Province, China.
| |
Collapse
|
31
|
Zhao S, Li X, Wang J, Wang H. The Role of the Effects of Autophagy on NLRP3 Inflammasome in Inflammatory Nervous System Diseases. Front Cell Dev Biol 2021; 9:657478. [PMID: 34079796 PMCID: PMC8166298 DOI: 10.3389/fcell.2021.657478] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a stable self-sustaining process in eukaryotic cells. In this process, pathogens, abnormal proteins, and organelles are encapsulated by a bilayer membrane to form autophagosomes, which are then transferred to lysosomes for degradation. Autophagy is involved in many physiological and pathological processes. Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome, containing NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and pro-caspase-1, can activate caspase-1 to induce pyroptosis and lead to the maturation and secretion of interleukin-1 β (IL-1 β) and IL-18. NLRP3 inflammasome is related to many diseases. In recent years, autophagy has been reported to play a vital role by regulating the NLRP3 inflammasome in inflammatory nervous system diseases. However, the related mechanisms are not completely clarified. In this review, we sum up recent research about the role of the effects of autophagy on NLRP3 inflammasome in Alzheimer’s disease, chronic cerebral hypoperfusion, Parkinson’s disease, depression, cerebral ischemia/reperfusion injury, early brain injury after subarachnoid hemorrhage, and experimental autoimmune encephalomyelitis and analyzed the related mechanism to provide theoretical reference for the future research of inflammatory neurological diseases.
Collapse
Affiliation(s)
- Shizhen Zhao
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Xiaotian Li
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jie Wang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Honggang Wang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
32
|
Chen J, Sun J, Hu Y, Wan X, Wang Y, Gao M, Liang J, Liu T, Sun X. MicroRNA-191-5p ameliorates amyloid-β 1-40 -mediated retinal pigment epithelium cell injury by suppressing the NLRP3 inflammasome pathway. FASEB J 2021; 35:e21184. [PMID: 33715208 DOI: 10.1096/fj.202000645rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022]
Abstract
Amyloid β (Aβ) is a crucial component of drusen, the hallmark of the early stage of age-related macular degeneration (AMD), and can cause retinal pigment epithelium (RPE) cell damage through activation of the inflammatory response. MicroRNAs play a critical role in inflammation. However, the mechanism underlying the effect of microRNAs on the NLRP3 inflammasome induced by Aβ remains poorly understood. In the present study, we demonstrated that Aβ1-40 -mediated RPE damage by inducing a decrease in endogenous miR-191-5p expression. This led to the upregulation of its target gene, C/EBPβ. C/EBPβ acts as a transcription factor for NLRP3, promotes its transcription, and upregulates the downstream inflammatory factors Caspase-1 and IL-1β. Correspondingly, overexpression of miR-191-5p alleviated RPE cell injury by suppressing inflammation. The present study elucidates a novel transcriptional regulatory mechanism of the NLRP3 inflammasome. Our findings suggest an anti-inflammatory effect of miR-191-5p in Aβ1-40 -induced RPE impairment, shedding light on novel preventive or therapeutic approaches for AMD-associated RPE impairment.
Collapse
Affiliation(s)
- Jieqiong Chen
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yifan Hu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yuwei Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Gao
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jian Liang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Lysosomal Functions in Glia Associated with Neurodegeneration. Biomolecules 2021; 11:biom11030400. [PMID: 33803137 PMCID: PMC7999372 DOI: 10.3390/biom11030400] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Lysosomes are cellular organelles that contain various acidic digestive enzymes. Despite their small size, they have multiple functions. Lysosomes remove or recycle unnecessary cell parts. They repair damaged cellular membranes by exocytosis. Lysosomes also sense cellular energy status and transmit signals to the nucleus. Glial cells are non-neuronal cells in the nervous system and have an active role in homeostatic support for neurons. In response to dynamic cues, glia use lysosomal pathways for the secretion and uptake of regulatory molecules, which affect the physiology of neighboring neurons. Therefore, functional aberration of glial lysosomes can trigger neuronal degeneration. Here, we review lysosomal functions in oligodendrocytes, astrocytes, and microglia, with emphasis on neurodegeneration.
Collapse
|
34
|
Feng YS, Tan ZX, Wu LY, Dong F, Zhang F. The involvement of NLRP3 inflammasome in the treatment of neurodegenerative diseases. Biomed Pharmacother 2021; 138:111428. [PMID: 33667787 DOI: 10.1016/j.biopha.2021.111428] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/06/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
In an ageing society, neurodegenerative diseases have attracted attention because of their high incidence worldwide. Despite extensive research, there is a lack of conclusive insights into the pathogenesis of neurodegenerative diseases, which limit the strategies for symptomatic treatment. Therefore, better elucidation of the molecular mechanisms involved in neurodegenerative diseases can provide an important theoretical basis for the discovery of new and effective prevention and treatment methods. The innate immune system is activated during the ageing process and in response to neurodegenerative diseases. Inflammasomes are multiprotein complexes that play an important role in the activation of the innate immune system. They mediate inflammatory reactions and pyroptosis, which are closely involved in neurodegeneration. There are different types of inflammasomes, although the nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome is the most common inflammasome; NLRP3 plays an important role in the pathogenesis of neurodegenerative diseases. In this review, we will discuss the mechanisms that are involved in the activation of the NLRP3 inflammasome and its crucial role in the pathology of neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. We will also review various treatments that target the NLRP3 inflammasome pathway and alleviate neuroinflammation. Finally, we will summarize the novel treatment strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lin-Yu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China; Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang 050051, PR China.
| |
Collapse
|
35
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
36
|
Feng YS, Tan ZX, Wu LY, Dong F, Zhang F. The involvement of NLRP3 inflammasome in the treatment of Alzheimer's disease. Ageing Res Rev 2020; 64:101192. [PMID: 33059089 DOI: 10.1016/j.arr.2020.101192] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/04/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and it is characterised by progressive deterioration in cognitive and memory abilities, which can severely influence the elderly population's daily living abilities. Although researchers have made great efforts in the field of AD, there are still no well-established strategies to prevent and treat this disease. Therefore, better clarification of the molecular mechanisms associated with the onset and progression of AD is critical to provide a theoretical basis for the establishment of novel preventive and therapeutic strategies. Currently, it is generally believed that neuroinflammation plays a key role in the pathogenesis of AD. Inflammasome, a multiprotein complex, is involved in the innate immune system, and it can mediate inflammatory responses and pyroptosis, which lead to neurodegeneration. Among the various types of inflammasomes, the NLRP3 inflammasome is the most characterised in neurodegenerative diseases, especially in AD. The activation of the NLRP3 inflammasome causes the generation of caspase-1-mediated interleukin (IL)-1β and IL-18 in microglia cells, where neuroinflammation is involved in the development and progression of AD. Thus, the NLRP3 inflammasome is likely to be a crucial therapeutic molecular target for AD via regulating neuroinflammation. In this review, we summarise the current knowledge on the role and regulatory mechanisms of the NLRP3 inflammasome in the pathogenic mechanisms of AD. We also focus on a series of potential therapeutic treatments targeting NLRP3 inflammasome for AD. Further clarification of the regulatory mechanisms of the NLRP3 inflammasome in AD may provide more useful clues to develop novel AD treatment strategies.
Collapse
|
37
|
Xu M, Yan T, Gong G, Wu B, He B, Du Y, Xiao F, Jia Y. Purification, structural characterization, and cognitive improvement activity of a polysaccharides from Schisandra chinensis. Int J Biol Macromol 2020; 163:497-507. [DOI: 10.1016/j.ijbiomac.2020.06.275] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 01/28/2023]
|
38
|
Ye A, Li W, Zhou L, Ao L, Fang W, Li Y. Targeting pyroptosis to regulate ischemic stroke injury: Molecular mechanisms and preclinical evidences. Brain Res Bull 2020; 165:146-160. [PMID: 33065175 DOI: 10.1016/j.brainresbull.2020.10.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
Abstract
Stroke is one of the leading causes of death worldwide with limited therapies. After ischemic stroke occurs, a robust sterile inflammatory response happens and lasts for days and determines neurological prognosis. Pyroptosis is an inflammatory programmed cell death characterized by cleavage of pore-forming proteins gasdermins as a result of activating caspases and inflammasomes. It has morphological characteristics of rapid plasma-membrane rupture and release of proinflammatory intracellular contents as well as cytokines. Recent researches implicate pyroptosis involvement in the pathogenesis of ischemic stroke and inhibition of pyroptosis attenuates ischemic brain injury. In this review, we discussed molecular mechanisms of pyroptosis, evidences for pyroptosis involvement in different kinds of the central nervous system cells, as well as potential inhibitors for intervention of pyroptosis. Based on the review, we hypothesize the feasibility of therapeutic strategies targeting pyroptosis in the context of ischemic stroke.
Collapse
Affiliation(s)
- Anqi Ye
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wanting Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lin Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Luyao Ao
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
39
|
Zhang Y, Zhao Y, Zhang J, Yang G. Mechanisms of NLRP3 Inflammasome Activation: Its Role in the Treatment of Alzheimer's Disease. Neurochem Res 2020; 45:2560-2572. [PMID: 32929691 DOI: 10.1007/s11064-020-03121-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease of progressive dementia which is characterized pathologically by extracellular neuritic plaques containing aggregated amyloid beta (Aβ) and intracellular hyperphosphorylated tau protein tangles in cerebrum. It has been confirmed that microglia-specific nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome-mediated chronic neuroinflammation plays a crucial role in the pathogenesis of AD. Stimulated by Aβ deposition, NLRP3 assembles and activates within microglia in the AD brain, leading to caspase-1 activation along with downstream interleukin (IL)-1β secretion, and subsequent inflammatory events. Activation of the NLRP3 inflammasome mediates microglia to exhibit inflammatory M1 phenotype, with high expression of caspase-1 and IL-1β. This leads to Aβ deposition and neuronal loss in the amyloid precursor protein (APP)/human presenilin-1 (PS1) mouse model of AD. However, NLRP3 or caspase-1 deletion in APP/PS1 mice promotes microglia to transform to an anti-inflammatory M2 phenotype, with decreased secretion of caspase-1 and IL-1β. It also results in improved cognition, enhanced Aβ clearance, and a lower cerebral inflammatory response. This result suggests that the NLRP3 inflammasome may be an appropriate target for reducing neuroinflammation and alleviating pathological processes in AD. In the present review, we summarize the generally accepted regulatory mechanisms of NLRP3 inflammasome activation, and explore its role in neuroinflammation. Furthermore, we speculate on the possible roles of microglia-specific NLRP3 activation in AD pathogenesis and consider potential therapeutic interventions targeting the NLRP3 inflammasome in AD.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Yuan Zhao
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Jian Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Guofeng Yang
- Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, People's Republic of China.
| |
Collapse
|
40
|
Sil S, Niu F, Chivero ET, Singh S, Periyasamy P, Buch S. Role of Inflammasomes in HIV-1 and Drug Abuse Mediated Neuroinflammaging. Cells 2020; 9:cells9081857. [PMID: 32784383 PMCID: PMC7464640 DOI: 10.3390/cells9081857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the effectiveness of combined antiretroviral therapy (cART) in suppressing virus replication, chronic inflammation remains one of the cardinal features intersecting HIV-1, cART, drug abuse, and likely contributes to the accelerated neurocognitive decline and aging in people living with HIV-1 (PLWH) that abuse drugs. It is also estimated that ~30–60% of PLWH on cART develop cognitive deficits associated with HIV-1-associated neurocognitive disorders (HAND), with symptomatology ranging from asymptomatic to mild, neurocognitive impairments. Adding further complexity to HAND is the comorbidity of drug abuse in PLWH involving activated immune responses and the release of neurotoxins, which, in turn, mediate neuroinflammation. Premature or accelerated aging is another feature of drug abusing PLWH on cART regimes. Emerging studies implicate the role of HIV-1/HIV-1 proteins, cART, and abused drugs in altering the inflammasome signaling in the central nervous system (CNS) cells. It is thus likely that exposure of these cells to HIV-1/HIV-1 proteins, cART, and/or abused drugs could have synergistic/additive effects on the activation of inflammasomes, in turn, leading to exacerbated neuroinflammation, ultimately resulting in premature aging referred to as “inflammaging” In this review, we summarize the current knowledge of inflammasome activation, neuroinflammation, and aging in central nervous system (CNS) cells such as microglia, astrocytes, and neurons in the context of HIV-1 and drug abuse.
Collapse
Affiliation(s)
| | | | | | | | | | - Shilpa Buch
- Correspondence: (P.P.); (S.B.); Tel.: +1-402-559-3165 (S.B.)
| |
Collapse
|
41
|
Lychee seed polyphenol inhibits Aβ-induced activation of NLRP3 inflammasome via the LRP1/AMPK mediated autophagy induction. Biomed Pharmacother 2020; 130:110575. [PMID: 32768883 DOI: 10.1016/j.biopha.2020.110575] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/14/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence indicates that the enhancement of microglial autophagy inhibits the NLRP3 inflammasome mediated neuroinflammation in Alzheimer's disease (AD). Meanwhile, low density lipoprotein receptor-related protein 1 (LRP1) highly expressed in microglia is able to negatively regulate neuroinflammation and positively regulate autophagy. In addition, we have previously reported that an active lychee seed fraction enriching polyphenol (LSP) exhibits anti-neuroinflammation in Aβ-induced BV-2 cells. However, its molecular mechanism of action is still unclear. In this study, we aim to investigate whether LSP inhibits the NLRP3 inflammasome mediated neuroinflammation and clarify its molecular mechanism in Aβ-induced BV-2 cells and APP/PS1 mice. The results showed that LSP dose- and time-dependently activated autophagy by increasing the expression of Beclin 1 and LC3II in BV-2 cells, which was regulated by the upregulation of LRP1 and its mediated AMPK signaling pathway. In addition, both the Western blotting and fluorescence microscopic results demonstrated that LSP could significantly suppress the activation of NLRP3 inflammasome by inhibiting the expression of NLRP3, ASC, the cleavage of caspase-1, and the release of IL-1β in Aβ(1-42)-induced BV-2 cells. In addition, the siRNA LRP1 successfully abolished the effect of LSP on the activation of AMPK and its mediated autophagy, as well as the inhibition of NLRP3 inflammasome. Furthermore, LSP rescued PC-12 cells which were induced by the conditioned medium from Aβ(1-42)-treated BV-2 cells. Moreover, LSP improved the cognitive function and inhibited the NLRP3 inflammasome in APP/PS1 mice. Taken together, LSP inhibited the NLRP3 inflammasome-mediated neuroinflammation in the in vitro and in vivo models of AD, which was closely associated with the LRP1/AMPK-mediated autophagy. Thus, the findings from this study further provide evidences for LSP serving as a potential drug for the treatment of AD in the future.
Collapse
|
42
|
Yang T, Feng X, Zhao Y, Zhang H, Cui H, Wei M, Yang H, Fan H. Dexmedetomidine Enhances Autophagy via α2-AR/AMPK/mTOR Pathway to Inhibit the Activation of NLRP3 Inflammasome and Subsequently Alleviates Lipopolysaccharide-Induced Acute Kidney Injury. Front Pharmacol 2020; 11:790. [PMID: 32670056 PMCID: PMC7326938 DOI: 10.3389/fphar.2020.00790] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background Acute kidney injury (AKI) is a severe complication of sepsis; however, no effective drugs have been found. Activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome is a major pathogenic mechanism of AKI induced by lipopolysaccharide (LPS). Autophagy, a process of intracellular degradation related to renal homeostasis, effectively restricts inflammatory responses. Herein, we explored the potential protective mechanisms of dexmedetomidine (DEX), which has confirmed anti-inflammatory effects, on LPS-induced AKI. Methods AKI was induced in rats by injecting 10 mg/kg of LPS intraperitoneally (i.p.). Wistar rats received intraperitoneal injections of DEX (30 µg/kg) 30 min before an intraperitoneal injection of LPS. Atipamezole (ATI) (250 µg/kg) and 3-methyladenine (3-MA) (15 mg/kg) were intraperitoneally injected 30 min before the DEX injection. Results DEX significantly attenuated renal injury. Furthermore, DEX decreased activation of the NLRP3 inflammasome and expression of interleukins 1β and 18. In addition, autophagy-related protein and gene analysis indicated that DEX could significantly enhance autophagy. Finally, we verified the pharmacological effects of DEX on the 5′-adenosine monophosphate-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) pathway. Atip and 3-MA significantly reversed the protective effects of DEX. Conclusions Our results suggest that the protective effects of DEX were mediated by enhanced autophagy via the α2-adrenoreceptor/AMPK/mTOR pathway, which decreased activation of the NLRP3 inflammasome. Above all, we verified the renal protective effects of DEX and offer a new treatment strategy for AKI.
Collapse
Affiliation(s)
- Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiujing Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haiyang Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hailin Cui
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mian Wei
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
43
|
Espinosa-Garcia C, Atif F, Yousuf S, Sayeed I, Neigh GN, Stein DG. Progesterone Attenuates Stress-Induced NLRP3 Inflammasome Activation and Enhances Autophagy following Ischemic Brain Injury. Int J Mol Sci 2020; 21:E3740. [PMID: 32466385 PMCID: PMC7312827 DOI: 10.3390/ijms21113740] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/18/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023] Open
Abstract
NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome inhibition and autophagy induction attenuate inflammation and improve outcome in rodent models of cerebral ischemia. However, the impact of chronic stress on NLRP3 inflammasome and autophagic response to ischemia remains unknown. Progesterone (PROG), a neuroprotective steroid, shows promise in reducing excessive inflammation associated with poor outcome in ischemic brain injury patients with comorbid conditions, including elevated stress. Stress primes microglia, mainly by the release of alarmins such as high-mobility group box-1 (HMGB1). HMGB1 activates the NLRP3 inflammasome, resulting in pro-inflammatory interleukin (IL)-1β production. In experiment 1, adult male Sprague-Dawley rats were exposed to social defeat stress for 8 days and then subjected to global ischemia by the 4-vessel occlusion model, a clinically relevant brain injury associated with cardiac arrest. PROG was administered 2 and 6 h after occlusion and then daily for 7 days. Animals were killed at 7 or 14 days post-ischemia. Here, we show that stress and global ischemia exert a synergistic effect in HMGB1 release, resulting in exacerbation of NLRP3 inflammasome activation and autophagy impairment in the hippocampus of ischemic animals. In experiment 2, an in vitro inflammasome assay, primary microglia isolated from neonatal brain tissue, were primed with lipopolysaccharide (LPS) and stimulated with adenosine triphosphate (ATP), displaying impaired autophagy and increased IL-1β production. In experiment 3, hippocampal microglia isolated from stressed and unstressed animals, were stimulated ex vivo with LPS, exhibiting similar changes than primary microglia. Treatment with PROG reduced HMGB1 release and NLRP3 inflammasome activation, and enhanced autophagy in stressed and unstressed ischemic animals. Pre-treatment with an autophagy inhibitor blocked Progesterone's (PROG's) beneficial effects in microglia. Our data suggest that modulation of microglial priming is one of the molecular mechanisms by which PROG ameliorates ischemic brain injury under stressful conditions.
Collapse
Affiliation(s)
- Claudia Espinosa-Garcia
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA; (F.A.); (S.Y.); (I.S.); (D.G.S.)
| | - Fahim Atif
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA; (F.A.); (S.Y.); (I.S.); (D.G.S.)
| | - Seema Yousuf
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA; (F.A.); (S.Y.); (I.S.); (D.G.S.)
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA; (F.A.); (S.Y.); (I.S.); (D.G.S.)
| | - Gretchen N. Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Donald G. Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA; (F.A.); (S.Y.); (I.S.); (D.G.S.)
| |
Collapse
|
44
|
Rong J, Xu J, Liu Q, Xu J, Mou T, Zhang X, Chi H, Zhou H. Anti-inflammatory effect of up-regulated microRNA-221-3p on coronary heart disease via suppressing NLRP3/ASC/pro-caspase-1 inflammasome pathway activation. Cell Cycle 2020; 19:1478-1491. [PMID: 32372677 DOI: 10.1080/15384101.2020.1754562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE As some evidence has demonstrated the role of microRNA-221 (miR-221) on coronary heart disease (CHD), the aim of the present study was to investigate the effect of miR-221-3p on CHD via regulating NLRP3/ASC/pro-caspase-1 inflammasome pathway. METHODS Sixty CHD patients and 60 healthy controls were collected to detect the expression of miR-221-3p, NLRP3, ASC, pro-caspase-1 in peripheral blood and the contents of related factors in serum. The rats model of CHD was injected with miR-221-3p agomir or miR-221-3p antagomir to explore its functions in miR-221-3p, NLRP3, ASC and pro-caspase-1 expression, electrocardiogram data, cardiomyocytes apoptosis, myocardial injury, inflammatory reaction and oxidative stress of CHD rats. RESULTS MiR-221-3p declined and NLRP3, ASC and pro-caspase-1 raised in CHD. Up-regulated miR-221-3p reduced the change value of J-point and T-wave, decreased NLRP3, ASC and pro-caspase-1 expression, suppressed apoptosis in cardiomyocytes, as well as suppressed myocardial injury, inflammatory reaction and oxidative stress in CHD rats. CONCLUSION This study highlights that up-regulated miR-221-3p suppresses the overactivation of NLRP3/ASC/pro-caspase-1 inflammasome pathway and has an anti-inflammatory effect in CHD. Thus, miR-221-3p may serve as a potential target for the treatment of CHD.
Collapse
Affiliation(s)
- Jingfeng Rong
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Jijie Xu
- Cardiovascular Medicine Institute, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Qian Liu
- Cardiovascular Medicine Institute, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Jianjun Xu
- Cardiothoracic Surgery Department, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Ting Mou
- Department of Cardiovascular, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Xuhua Zhang
- Department of Cardiovascular, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Hao Chi
- Cardiothoracic Surgery Department, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| | - Hua Zhou
- Department of Cardiovascular, Shuguang Hospital Affiliated to Shanghai University of Chinese Medicine , Shanghai, China
| |
Collapse
|
45
|
Xia S, Ni Y, Zhou Q, Liu H, Xiang H, Sui H, Shang D. Emodin Attenuates Severe Acute Pancreatitis via Antioxidant and Anti-inflammatory Activity. Inflammation 2020; 42:2129-2138. [PMID: 31605249 DOI: 10.1007/s10753-019-01077-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
There is no specific drug to treat severe acute pancreatitis (SAP), which induces substantial medical and social burden. Many studies have reported the beneficial effects of emodin against SAP in vivo and in vitro. However, the underlying mechanism has been unclear. This paper described the design and implementation of anti-inflammatory and antioxidant activity of emodin. Emodin restored the pathological damage of SAP and simultaneously decreased the high levels of serum amylase, lipase, TNF-α, and IL-18 in the peripheral blood of SAP rat. Emodin reversed reactive oxygen species (ROS) in neutrophils derived from SAP rat. The levels of voltage-dependent anion channel 1 (VDAC1), NOD-like receptor protein 3 (NLRP3), caspase-1, and IL-18 were examined to analyze the change of inflammasome-related mediators between SAP and emodin treatment. These findings suggest that emodin plays its protective role on SAP against oxidative stress and inflammasome signals.
Collapse
Affiliation(s)
- Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, China
| | - Yujia Ni
- Department of Geriatrics, Shaoxing People's Hospital, No. 568, Zhongxing North Road, Shaoxing, Zhejiang, China
| | - Qi Zhou
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Han Liu
- Department of Oral Pathology, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Hong Xiang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, China
| | - Hua Sui
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Dong Shang
- Institute (College) of Integrative Medicine, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China.
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, China.
| |
Collapse
|