1
|
Asadi Y, Moundounga RK, Chakroborty A, Pokokiri A, Wang H. FOXOs and their roles in acute and chronic neurological disorders. Front Mol Biosci 2025; 12:1538472. [PMID: 40260403 PMCID: PMC12010098 DOI: 10.3389/fmolb.2025.1538472] [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: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 04/23/2025] Open
Abstract
The forkhead family of transcription factors of class O (FOXOs) consisting of four functionally related proteins, FOXO1, FOXO3, FOXO4, and FOXO6, are mammalian homologs of daf-16 in Caenorhabditis elegans and were previously identified as tumor suppressors, oxidative stress sensors, and cell survival modulators. Under normal physiological conditions, FOXO protein activities are negatively regulated by phosphorylation via the phosphoinositide 3-kinase (PI3K)-Akt pathway, a well-known cell survival pathway: Akt phosphorylates FOXOs to inactivate their transcriptional activity by relocalizing FOXOs from the nucleus to the cytoplasm for degradation. However, under oxidative stress or absent the cellular survival drive of growth factors, FOXO proteins translocate to the nucleus and upregulate a series of target genes, thereby promoting cell growth arrest and cell death and altering mitochondrial homeostasis. FOXO gene expression is also regulated by other transcriptional factors such as p53 or autoregulation by their activities and end products. Here we summarize the structure, posttranslational modifications, and translocation of FOXOs linking to their transcriptional control of cellular functions, survival, and death, emphasizing their role in regulating the cellular response to some acute insults and chronic neurological disorders. This review will conclude with a brief section on potential therapeutic interventions that can be used to modulate FOXOs' activities when treating acute and chronic neurological disorders.
Collapse
Affiliation(s)
- Yasin Asadi
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rozenn K. Moundounga
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Anand Chakroborty
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Augustina Pokokiri
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hongmin Wang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
2
|
Ravanidis S, Bougea A, Koros C, Simitsi AM, Kokotis P, Stefanis L, Doxakis E. Plasma miRNA Biomarker Signatures in Parkinsonian Syndromes. Mol Neurobiol 2025:10.1007/s12035-025-04890-w. [PMID: 40184025 DOI: 10.1007/s12035-025-04890-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Diagnosing atypical parkinsonian syndromes (APS) remains challenging due to overlapping clinical features and limited diagnostic tools. Brain-enriched microRNAs (miRNAs), which regulate neuronal development and function, are detectable in plasma and could serve as molecular biomarkers. This prospective study aimed to identify plasma brain-enriched miRNAs that can distinguish APS and elucidate affected molecular pathways. Reverse transcription-quantitative PCR (RT-qPCR) was performed on plasma samples from patients with idiopathic Parkinson's disease (iPD), multiple system atrophy (MSA), including the cerebellar subtype (MSA-C) and the parkinsonian subtype (MSA-P), progressive supranuclear palsy (PSP), and healthy controls. MiRNA expression analysis revealed distinct molecular fingerprints for each parkinsonian syndrome, with opposite trends between MSA and iPD compared to controls, suggesting distinct pathogenic mechanisms. Most dysregulated miRNAs clustered at chromosome (Chr)14q32 and shared binding sites for CREB1, CEBPB, and MAZ transcription factors. Pathway analysis revealed enrichment in prion diseases, Hippo signaling, TGF-beta signaling, and FoxO signaling pathways.
Collapse
Affiliation(s)
- Stylianos Ravanidis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece
| | - Anastasia Bougea
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, 11528, Athens, Greece
| | - Christos Koros
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, 11528, Athens, Greece
| | - Athina-Maria Simitsi
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, 11528, Athens, Greece
| | - Panagiotis Kokotis
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, 11528, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical Research, Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece
- First Department of Neurology, National and Kapodistrian University of Athens Medical School, 11528, Athens, Greece
| | - Epaminondas Doxakis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece.
| |
Collapse
|
3
|
Nisar A, Khan S, Pan Y, Hu L, Yang P, Gold NM, Zhou Z, Yuan S, Zi M, Mehmood SA, He Y. The Role of Hypoxia in Longevity. Aging Dis 2025:AD.2024.1630. [PMID: 39965249 DOI: 10.14336/ad.2024.1630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/15/2025] [Indexed: 02/20/2025] Open
Abstract
Aging is marked by a progressive decrease in physiological function and reserve capacity, which results in increased susceptibility to diseases. Understanding the mechanisms of driving aging is crucial for extending health span and promoting human longevity. Hypoxia, marked by reduced oxygen availability, has emerged as a promising area of study within aging research. This review explores recent findings on the potential of oxygen restriction to promote healthy aging and extend lifespan. While the role of hypoxia-inducible factor 1 (HIF-1) in cellular responses to hypoxia is well-established, its impact on lifespan remains complex and context-dependent. Investigations in invertebrate models suggest a role for HIF-1 in longevity, while evidence in mammalian models is limited. Hypoxia extends the lifespan independent of dietary restriction (DR), a known intervention underlying longevity. However, both hypoxia and DR converge on common downstream effectors, such as forkhead box O (FOXO) and flavin-containing monooxygenase (FMOs) to modulate the lifespan. Further work is required to elucidate the molecular mechanisms underlying hypoxia-induced longevity and optimize clinical applications. Understanding the crosstalk between HIF-1 and other longevity-associated pathways is crucial for developing interventions to enhance lifespan and healthspan. Future studies may uncover novel therapeutic strategies to promote healthy aging and longevity in human populations.
Collapse
Affiliation(s)
- Ayesha Nisar
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Sawar Khan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410083, China
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan
| | - Yongzhang Pan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Li Hu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Pengyun Yang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Naheemat Modupeola Gold
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Zhen Zhou
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Shengjie Yuan
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Meiting Zi
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | | | - Yonghan He
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| |
Collapse
|
4
|
Cui Y, Bai M, Gao S, Zhao H, Mei X. Zinc ions facilitate metabolic bioenergetic recovery post spinal cord injury by activating microglial mitophagy through the STAT3-FOXO3a-SOD2 pathway. Free Radic Biol Med 2025; 227:64-79. [PMID: 39613048 DOI: 10.1016/j.freeradbiomed.2024.11.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Spinal cord injury (SCI) is a devastating condition of the central nervous system (CNS) with high global rates of disability and mortality, and no effective cure currently available. Microglia play a critical role in the progression of SCI, and enhancing their metabolic function may facilitate tissue repair and recovery. Mitochondrial dysfunction is a key feature of metabolic impairment, with the regulation of autophagy being essential for maintaining mitochondrial homeostasis and cell survival. The transcription factor Forkhead box O3a (FOXO3a) is integral to cellular metabolism, mitochondrial dysfunction, and oxidative stress responses, yet its role in post-SCI microglial metabolism remains underexplored. In this study, single-cell RNA sequencing reveals the crucial involvement of the FOXO signaling pathway in zinc ion-mediated enhancement of microglial metabolism. Mechanistically, oxidative stress-induced reactive oxygen species (ROS) accumulation exacerbates metabolic dysfunction by promoting excessive mitochondrial fission and impairing mitophagy. Importantly, zinc ions induce the nuclear translocation of FOXO3a, leading to its activation as a transcription factor. This activation enhances mitochondrial autophagy and fusion processes, thereby restoring microglial metabolic capacity. Our findings suggest that the zinc ion regulation of the STAT3-FOXO3a-SOD2 axis is pivotal in modulating mitochondrial gene expression, which governs microglial energy homeostasis and improves the spinal cord microenvironment, potentially enhancing neuronal survival. These insights highlight a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yang Cui
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, China
| | - Mingyu Bai
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, China
| | - Shuang Gao
- Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou, Liaoning Province, 121000, China
| | - Haosen Zhao
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, China; Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou, Liaoning Province, 121000, China; Liaoning Provincial Clinical Research Center for Bone Tissue Engineering, Jinzhou, Liaoning Province, 121000, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou, Liaoning Province, 121000, China.
| | - Xifan Mei
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, China; Liaoning Provincial Key Laboratory of Medical Tissue Engineering, Jinzhou, Liaoning Province, 121000, China; Liaoning Provincial Clinical Research Center for Bone Tissue Engineering, Jinzhou, Liaoning Province, 121000, China; Liaoning Provincial Collaborative Innovation Center of Medical Testing and Drug Development, Jinzhou, Liaoning Province, 121000, China.
| |
Collapse
|
5
|
Lee JH, Lee KH, Ryu JH, Kim MJ, Kim E, Lee SY, Han SC, Choi BT, Shin YI, Shin HK. LM22A-4-loaded smart mesoporous balls enhance neuroprotection and functional recovery after ischemic stroke. Biomed Pharmacother 2025; 183:117863. [PMID: 39842267 DOI: 10.1016/j.biopha.2025.117863] [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: 09/26/2024] [Revised: 12/23/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025] Open
Abstract
Stroke is globally recognized as the second leading cause of death, significantly impairing both motor and cognitive functions. Enhancing regeneration after stroke is crucial for restoring these functions and necessitates strategies to promote neuroregeneration to achieve better post-stroke outcomes. Brain-derived neurotrophic factor (BDNF) plays a key role in neuroregeneration by influencing motor ability, learning, memory, and rehabilitation after stroke. However, challenges such as the substantial protein size, short half-life of BDNF, and blood-brain barrier hinder its efficient delivery to the brain. In this study, LM22A-4, a BDNF mimetic, was utilized and delivered through a Smart Mesoporous Ball (SMB-3) system to target the ischemic injured brain and explore its potential therapeutic effects in a mouse ischemic stroke model. Treatment with LM22A-4-loaded SMB-3 (LM22A-4-SMB-3) markedly restored neurological, motor, and cognitive deficits following ischemic stroke compared to LM22A-4 alone. Additionally, administration of LM22A-4-SMB-3 reduced apoptotic cell death and glial activation, as evidenced by the TUNEL assay results, and decreased GFAP and Iba-1 expression levels. Furthermore, the phosphorylation of TrkB and Akt, but not that of Erk, was considerably increased in the LM22A-4-SMB-3-treated group. Treatment also enhanced the number of BrdU+/NeuN+ cells, with a marked reduction in post-stroke brain atrophy. These findings suggest that LM22A-4-SMB-3 can attenuate ischemic damage and recover neurological, motor, and cognitive functions by increasing p-TrkB and p-Akt levels and promoting neurogenesis. Therefore, SMB-3-mediated delivery of LM22A-4 presents a potentially applicable delivery system, and LM22A-4-SMB-3 use could be considered a novel therapeutic strategy to improve post-stroke outcomes.
Collapse
Affiliation(s)
- Jae Ho Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Kyeong Hyeon Lee
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Ji Hyeon Ryu
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Min Jae Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Eunji Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Seo-Yeon Lee
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Sang-Cheol Han
- CEN Co., Ltd. Nano-Convergence Center, 761 Muan-ro, Miryang 50404, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Yong-Il Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea.
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea.
| |
Collapse
|
6
|
Dong SS, Li MY, Yu XP, Kan YN, Dai XH, Zheng L, Cao HT, Duan WH, Luo EL, Zou W. Baihui-Penetrating-Qubin Acupuncture Attenuates Neurological Deficits Through SIRT1/FOXO1 Reducing Oxidative Stress and Neuronal Apoptosis in Intracerebral Hemorrhage Rats. Brain Behav 2024; 14:e70095. [PMID: 39682063 DOI: 10.1002/brb3.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a significant global disease with high mortality and disability. As of now, there is no effective therapy available. Oxidative stress and neuronal apoptosis play essential roles in ICH, determining neuronal survival. In our preliminary studies, we found that Baihui-penetrating-Qubin acupuncture could improve neurological deficits and neuropathological damage in the perihematomal area in ICH rats. The SIRT1/FOXO1 signaling pathway has been reported to mediate antioxidant and anti-neuronal apoptosis. This study aimed to investigate the effects of Baihui-penetrating-Qubin acupuncture on oxidative stress and neuronal apoptosis after ICH and the role of SIRT1/FOXO1 in acupuncture's neuroprotection. METHODS ICH rat models were established by autologous tail blood (50 µL) infusion into the caudate nucleus. EX527, SIRT1-specific inhibitor was intraperitoneally administered 3 days before ICH. Baihui-penetrating-Qubin acupuncture treatment was performed once a day for 30 min after ICH. Neurological deficits were evaluated using the modified neurological severity score (mNSS). Brain edema was evaluated using brain water content. HE staining and Nissl staining were used to evaluate neuropathological damage in the perihematomal area. Terminal deoxynucleotidyl transferase dUTP nick end labeling was used to quantify neuronal apoptosis. Specific kits were used to detect the levels of SOD, CAT, GSH-Px in the brain. The oxidative DNA damage was evaluated using enzyme-linked immunosorbent assay to detect the level of 8-hydroxyguanosine (8-OHdG). Western blot was used to evaluate the expressions of SIRT1, Ac-FOXO1, FOXO1, Bcl-2, and Bax. Immunofluorescence staining was conducted to detect the cellular localization of SIRT1. RESULTS Baihui-penetrating-Qubin acupuncture improved the neurological deficits and brain edema, reduced the pathological injury and neuronal degeneration in 3 days in the perihematomal area after ICH. Mechanistically, acupuncture reduced oxidative stress injury and neuronal apoptosis via activating SIRT1/FOXO1 pathway. The neuroprotective effects of acupuncture were abolished by injection of the SIRT1 inhibitor EX527. CONCLUSIONS Baihui-penetrating-Qubin acupuncture could reduce oxidative stress and neuronal apoptosis, at least in part, through the SIRT1/FOXO1 signaling pathway, improving neurological deficits and neuropathological damage after ICH. These findings suggest that Baihui-penetrating-Qubin acupuncture is an effective therapy for ICH, as well as targeting SIRT1 signaling to promote neuron survival could be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Shan-Shan Dong
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
- Second Department of Traditional Chinese Medicine, Medical School, South China Hospital, Shenzhen University, Shenzhen, P. R. China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Ming-Yue Li
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| | - Xue-Ping Yu
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| | - Yu-Na Kan
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, P. R. China
| | - Xiao-Hong Dai
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| | - Lei Zheng
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| | - Hong-Tao Cao
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| | - Wen-Hui Duan
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| | - En-Li Luo
- Second Department of Traditional Chinese Medicine, Medical School, South China Hospital, Shenzhen University, Shenzhen, P. R. China
| | - Wei Zou
- Acupuncture and Moxibustion Department, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, P. R. China
| |
Collapse
|
7
|
Du X, Zhang M, Wang R, Zeng Z, Zhao W, Fang B, Lan H, Hung W, Gao H. Bifidobacterium lactis-Derived Vesicles Attenuate Hippocampal Neuroinflammation by Targeting IL-33 to Regulate FoxO6/P53 Signaling. Nutrients 2024; 16:3586. [PMID: 39519420 PMCID: PMC11547434 DOI: 10.3390/nu16213586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Hippocampal Neuroinflammation (HNF) is a critical driver of cognitive impairment. The lipopolysaccharide (LPS) accumulate amyloid beta (Aβ) and lead to HNF. The Bifidobacterium lactis (BL) 99 have anti-inflammatory ability. However, whether BL99-derived microbiota-derived vesicles (MV) could alleviate LPS-induced HNF remains unclear. METHODS To investigate, we used ultrafiltration with ultracentrifuge to extract BL99-derived-MV (BL99-MV). We used hippocampal neuronal HT22 cells (HT22) to establish the LPS-induced HNF model, and explored whether BL99-MV alleviate LPS-induced HNF. RESULTS The confocal microscopy showed that BL99-MV were taken up by HT22 and reduced the oxidative stress (ROS) level. The PCR showed that BL99-MV up-regulate IL-10 level, and down-regulate TNF-α, IL-1β, and IL-6. Transcriptomic analysis revealed 4127 differentially expressed genes, with 2549 genes upregulated and 1578 genes downregulated in the BL99-MV group compared to the LPS group. Compared to the LPS group, BL99-MV decreased FoxO6, IL-33, P53, and NFκB expression, but increased FoxO1 and Bcl2 expression. The WB showed that BL99-MV modulated NFκB, FoxO6, P53, Caspase9, and Caspase3 protein expression by reducing IL-33 expression in HT22. The findings demonstrated IL-33 as a regulator for FoxO6/P53 signaling. CONCLUSIONS Here, we hypothesized that BL99-MV alleviated LPS-induced HNF to promote HT22 survival and synaptic development by regulating FoxO6/P53 signaling by targeting IL-33.
Collapse
Affiliation(s)
- Xiaoyu Du
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (X.D.); (M.Z.)
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (X.D.); (M.Z.)
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.W.); (W.Z.); (B.F.)
| | - Zhaozhong Zeng
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; (Z.Z.); (H.L.)
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010100, China
| | - Wen Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.W.); (W.Z.); (B.F.)
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.W.); (W.Z.); (B.F.)
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; (Z.Z.); (H.L.)
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010100, China
| | - Weilian Hung
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; (Z.Z.); (H.L.)
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010100, China
| | - Haina Gao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (X.D.); (M.Z.)
| |
Collapse
|
8
|
Giunti S, Blanco MG, De Rosa MJ, Rayes D. The ketone body β-hydroxybutyrate ameliorates neurodevelopmental deficits in the GABAergic system of daf-18/PTEN Caenorhabditis elegans mutants. eLife 2024; 13:RP94520. [PMID: 39422188 PMCID: PMC11488850 DOI: 10.7554/elife.94520] [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] [Indexed: 10/19/2024] Open
Abstract
A finely tuned balance between excitation and inhibition (E/I) is essential for proper brain function. Disruptions in the GABAergic system, which alter this equilibrium, are a common feature in various types of neurological disorders, including autism spectrum disorders (ASDs). Mutations in Phosphatase and Tensin Homolog (PTEN), the main negative regulator of the phosphatidylinositol 3-phosphate kinase/Akt pathway, are strongly associated with ASD. However, it is unclear whether PTEN deficiencies can differentially affect inhibitory and excitatory signaling. Using the Caenorhabditis elegans neuromuscular system, where both excitatory (cholinergic) and inhibitory (GABAergic) inputs regulate muscle activity, we found that daf-18/PTEN mutations impact GABAergic (but not cholinergic) neurodevelopment and function. This selective impact results in a deficiency in inhibitory signaling. The defects observed in the GABAergic system in daf-18/PTEN mutants are due to reduced activity of DAF-16/FOXO during development. Ketogenic diets (KGDs) have proven effective for disorders associated with E/I imbalances. However, the mechanisms underlying their action remain largely elusive. We found that a diet enriched with the ketone body β-hydroxybutyrate during early development induces DAF-16/FOXO activity, therefore improving GABAergic neurodevelopment and function in daf-18/PTEN mutants. Our study provides valuable insights into the link between PTEN mutations and neurodevelopmental defects and delves into the mechanisms underlying the potential therapeutic effects of KGDs.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - María Gabriela Blanco
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| |
Collapse
|
9
|
Kathanadan Chackochan B, Johnson S, Thameemul Ansari HJ, Vengellur A, Sivan U, Koyyappurath S, P S BC. Transcriptomic analysis of CNTF-treated mouse subventricular zone-derived neurosphere culture reveals key transcription factor genes related to adult neurogenesis. Heliyon 2024; 10:e38496. [PMID: 39430537 PMCID: PMC11490819 DOI: 10.1016/j.heliyon.2024.e38496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/03/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Neural Stem Progenitor Cells (NSPCs) maintenance and neuronal cell differentiation are the two key aspects of sustained neurogenesis in the adult mammalian brain. Transcription factors (TFs) are known to regulate these biological processes under the influence of various neurotrophic factors. Understanding the role of key TF genes in regulating adult neurogenesis is essential for determining the functional complexity and neuronal diversity seen in the adult mammalian brain. Although several molecular mechanisms leading to adult neurogenesis have been reported, details on its transcriptional regulation are still limited. Our initial results showed that Ciliary Neurotrophic Factor (CNTF) induced neuronal differentiation in SVZ-derived NSPC cultures. To investigate further the role of CNTF in inducing the expression of TF genes related to adult neurogenesis and the potential pathways involved, whole transcriptome RNA-sequencing (RNA-seq) analysis was done in CNTF-treated Sub-ventricular Zone derived neurosphere cultures from the mouse brain. The study revealed 483 differentially expressed genes (DEGs), among which 33 DEGs were identified as coding for transcription factors (TFs). Kyoto Encyclopedia of Gene and Genomes (KEGG) analysis revealed MAPK, PI3K-Akt, and FoxO as the significantly enriched signaling pathways. Gene co-expression network analysis identified five upregulated TF genes related to adult neurogenesis (Runx1, Hmga2, Fos, ID2, and Prrx1) in a single cluster, interacting with each other, and was also validated by quantitative PCR. Our data suggest several potential TFs that may act as critical regulators in the intrinsic transcriptional networks driving the adult neurogenesis process. Further investigation into these molecular regulators may yield a homogeneous population of neuronal progenitors for translational stem cell studies in the future.
Collapse
Affiliation(s)
- Bins Kathanadan Chackochan
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Sinoy Johnson
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Hilmi Jaufer Thameemul Ansari
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ajith Vengellur
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Unnikrishnan Sivan
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Kerala University of Fisheries and Ocean Studies, Cochin -682506, Kerala, India
| | - Sayuj Koyyappurath
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Baby Chakrapani P S
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Excellence in Neurodegeneration and Brain Health, Kerala, India
| |
Collapse
|
10
|
Ning C, Jin M, Cai Y, Fan L, Hu K, Lu Z, Zhang M, Chen C, Li Y, Hu N, Zhang D, Liu Y, Chen S, Jiang Y, He C, Wang Z, Cao Z, Li H, Li G, Ma Q, Geng H, Tian W, Zhang H, Yang X, Huang C, Wei Y, Li B, Zhu Y, Li X, Miao X, Tian J. Genetic architectures of the human hippocampus and those involved in neuropsychiatric traits. BMC Med 2024; 22:456. [PMID: 39394562 PMCID: PMC11470718 DOI: 10.1186/s12916-024-03682-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND The hippocampus, with its complex subfields, is linked to numerous neuropsychiatric traits. While most research has focused on its global structure or a few specific subfields, a comprehensive analysis of hippocampal substructures and their genetic correlations across a wide range of neuropsychiatric traits remains underexplored. Given the hippocampus's high heritability, considering hippocampal and subfield volumes (HASV) as endophenotypes for neuropsychiatric conditions is essential. METHODS We analyzed MRI-derived volumetric data of hippocampal and subfield structures from 41,525 UK Biobank participants. Genome-wide association studies (GWAS) on 24 HASV traits were conducted, followed by genetic correlation, overlap, and Mendelian randomization (MR) analyses with 10 common neuropsychiatric traits. Polygenic risk scores (PRS) based on HASV traits were also evaluated for predicting these traits. RESULTS Our analysis identified 352 independent genetic variants surpassing a significance threshold of 2.1 × 10-9 within the 24 HASV traits, located across 93 chromosomal regions. Notably, the regions 12q14.3, 17q21.31, 12q24.22, 6q21, 9q33.1, 6q25.1, and 2q24.2 were found to influence multiple HASVs. Gene set analysis revealed enrichment of neural differentiation and signaling pathways, as well as protein binding and degradation. Of 240 HASV-neuropsychiatric trait pairs, 75 demonstrated significant genetic correlations (P < 0.05/240), revealing 433 pleiotropic loci. Particularly, genes like ACBD4, ARHGAP27, KANSL1, MAPT, ARL17A, and ARL17B were involved in over 50 HASV-neuropsychiatric pairs. Leveraging Mendelian randomization analysis, we further confirmed that atrophy in the left hippocampus, right hippocampus, right hippocampal body, and right CA1-3 region were associated with an increased risk of developing Parkinson's disease (PD). Furthermore, PRS for all four HASVs were significantly linked to a higher risk of Parkinson's disease (PD), with the highest hazard ratio (HR) of 1.30 (95% CI 1.18-1.43, P = 6.15 × 10⁻⁸) for right hippocampal volume. CONCLUSIONS These findings highlight the extensive distribution of pleiotropic genetic determinants between HASVs and neuropsychiatric traits. Moreover, they suggest a significant potential for effectively managing and intervening in these diseases during their early stages.
Collapse
Affiliation(s)
- Caibo Ning
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Meng Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yimin Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Linyun Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Kexin Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Can Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yanmin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Naifan Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Donghui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yizhuo Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Shuoni Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yuan Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Chunyi He
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zhuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Zilong Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Hanting Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Gaoyuan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Qianying Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Hui Geng
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Wen Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Heng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Xiaojun Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chaoqun Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yongchang Wei
- Department of Gastrointestinal Oncology, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China
| | - Xiangpan Li
- Department of Radiation Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China.
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China.
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan, 430071, China.
- Department of Oncology, Renmin Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
11
|
Peng S, Xu Y, Lin X. Inhibition of FOXO3 ameliorates ropivacaine-induced nerve cell damage through the miR-126-5p/TRAF6 axis. In Vitro Cell Dev Biol Anim 2024; 60:1109-1120. [PMID: 39227495 DOI: 10.1007/s11626-024-00970-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
Local anesthetics, such as ropivacaine (Ropi), are toxic to nerve cells. We aimed to explore the role of forkhead box O3 (FOXO3) in Ropi-induced nerve injury to provide a theoretical basis for reducing the anesthetic neurotoxicity. SK-N-SH cells were cultured and treated with different concentrations of Ropi. Cell viability, apoptosis, cytotoxicity (LDH/ROS/SOD), and levels of FOXO3, miR-126-5p, and tumor necrosis factor receptor-associated factor 6 (TRAF6) were detected. The enrichment of FOXO3 on the miR-126-5p promoter was analyzed. The binding relationships among FOXO3, miR-126-5p promoter sequence, and TRAF6 3'UTR sequence were verified. Combined experiments detected the regulatory role of FOXO3/miR-126-5p/TRAF6 in Ropi-induced nerve injury. FOXO3 was upregulated in Ropi-induced nerve cell damage. Inhibition of FOXO3 ameliorated Ropi-induced decreased cell viability, and increased apoptosis and cytotoxicity. FOXO3 bound to the miR-126-5p promoter and inhibited its expression, thereby counteracting miR-126-5p-induced repression. miR-126-5p inhibition and TRAF6 overexpression partially reversed the alleviative effect of FOXO3 inhibition on Ropi-induced nerve cell damage. In conclusion, FOXO3 aggravated the neurotoxicity of Ropi through miR-126-5p downregulation and TRAF6 upregulation, suggesting that FOXO3 inhibitor could be an adjuvant agent for local anesthetics, to alleviate local anesthetics-induced neurotoxicity.
Collapse
Affiliation(s)
- Song Peng
- Department of Anesthesiology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yuzeng Xu
- Department of Anesthesiology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xiao Lin
- Department of Anesthesiology, Women's Hospital School of Medicine Zhejiang University, 1 Bachelor Road, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
12
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
13
|
Maalouly G, Martin CMA, Baz Y, Saliba Y, Baramili AM, Fares N. Antioxidant and Anti-Apoptotic Neuroprotective Effects of Cinnamon in Imiquimod-Induced Lupus. Antioxidants (Basel) 2024; 13:880. [PMID: 39061948 PMCID: PMC11274315 DOI: 10.3390/antiox13070880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Despite accumulating evidence correlating oxidative stress with lupus disease activity, the brain redox pathways are still poorly investigated. Cinnamomum cassia, a widely used spice with powerful antioxidant properties, could be a novel therapeutic candidate in lupus. METHODS C57BL/6J female mice were divided into five groups: sham, sham-cinnamon, lupus, lupus-cinnamon starting from induction, and lupus-cinnamon starting two weeks before induction. Lupus was induced by skin application on the right ear with 1.25 mg of 5% imiquimod cream three times per week for six weeks. Cinnamomum cassia was given orally, five days per week, at 200 mg/kg. RESULTS Concomitant to TLR7-MYD88 pathway activation, the p-NRF2/NRF2 and p-FOXO3/FOXO3 ratios were increased in the hippocampus and alleviated by cinnamon treatment. BCL-2 positivity was enhanced in hippocampal neurons and reversed only by preventive cinnamon administration. In vitro, exposure of hippocampal cells to the plasma of different groups induced a surge in oxidative stress. This was associated with an increased t-BID/BID ratio. Cinnamon treatment, particularly in the preventive arm, normalized these modifications. CONCLUSIONS Our study shows a neuroprotective effect of cinnamon by rescuing brain redox and apoptosis homeostasis in lupus, paving the way for its use as a natural therapeutic compound in the clinical management of lupus.
Collapse
Affiliation(s)
| | | | | | | | | | - Nassim Fares
- Laboratory of Research in Physiology and Pathophysiology, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University of Beirut, Beirut 1104 2020, Lebanon; (G.M.); (C.-M.-A.M.); (Y.B.); (Y.S.); (A.-M.B.)
| |
Collapse
|
14
|
Johnson GA, Kodati B, Nahomi RB, Pham JH, Krishnamoorthy VR, Phillips NR, Krishnamoorthy RR, Nagaraj RH, Stankowska DL. Mechanisms contributing to inhibition of retinal ganglion cell death by cell permeable peptain-1 under glaucomatous stress. Cell Death Discov 2024; 10:305. [PMID: 38942762 PMCID: PMC11213865 DOI: 10.1038/s41420-024-02070-8] [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: 02/22/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/30/2024] Open
Abstract
This study assesses the neuroprotective potential of CPP-P1, a conjugate of an anti-apoptotic peptain-1 (P1) and a cell-penetrating peptide (CPP) in in vitro, in vivo, and ex vivo glaucoma models. Primary retinal ganglion cells (RGCs) were subjected to either neurotrophic factor (NF) deprivation for 48 h or endothelin-3 (ET-3) treatment for 24 h and received either CPP-P1 or vehicle. RGC survival was analyzed using a Live/Dead assay. Axotomized human retinal explants were treated with CPP-P1 or vehicle for seven days, stained with RGC marker RBPMS, and RGC survival was analyzed. Brown Norway (BN) rats with elevated intraocular pressure (IOP) received weekly intravitreal injections of CPP-P1 or vehicle for six weeks. RGC function was evaluated using a pattern electroretinogram (PERG). RGC and axonal damage were also assessed. RGCs from ocular hypertensive rats treated with CPP-P1 or vehicle for seven days were isolated for transcriptomic analysis. RGCs subjected to 48 h of NF deprivation were used for qPCR target confirmation. NF deprivation led to a significant loss of RGCs, which was markedly reduced by CPP-P1 treatment. CPP-P1 also decreased ET-3-mediated RGC death. In ex vivo human retinal explants, CPP-P1 decreased RGC loss. IOP elevation resulted in significant RGC loss in mid-peripheral and peripheral retinas compared to that in naive rats, which was significantly reduced by CPP-P1 treatment. PERG amplitude decline in IOP-elevated rats was mitigated by CPP-P1 treatment. Following IOP elevation in BN rats, the transcriptomic analysis showed over 6,000 differentially expressed genes in the CPP-P1 group compared to the vehicle-treated group. Upregulated pathways included CREB signaling and synaptogenesis. A significant increase in Creb1 mRNA and elevated phosphorylated Creb were observed in CPP-P1-treated RGCs. Our study showed that CPP-P1 is neuroprotective through CREB signaling enhancement in several settings that mimic glaucomatous conditions. The findings from this study are significant as they address the pressing need for the development of efficacious therapeutic strategies to maintain RGC viability and functionality associated with glaucoma.
Collapse
Affiliation(s)
- Gretchen A Johnson
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Bindu Kodati
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology and Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Rooban B Nahomi
- Department of Ophthalmology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Jennifer H Pham
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | - Nicole R Phillips
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Raghu R Krishnamoorthy
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology and Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Ram H Nagaraj
- Department of Ophthalmology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Dorota L Stankowska
- North Texas Eye Research Institute, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA.
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
15
|
Wei W, Ma S, Fu B, Song R, Guo H. Human-specific insights into candidate genes and boosted discoveries of novel loci illuminate roles of neuroglia in reading disorders. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12899. [PMID: 38752599 PMCID: PMC11097622 DOI: 10.1111/gbb.12899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
Reading disorders (RD) are human-specific neuropsychological conditions associated with decoding printed words and/or reading comprehension. So far only a handful of candidate genes segregated in families and 42 loci from genome-wide association study (GWAS) have been identified that jointly provided little clues of pathophysiology. Leveraging human-specific genomic information, we critically assessed the RD candidates for the first time and found substantial human-specific features within. The GWAS candidates (i.e., population signals) were distinct from the familial counterparts and were more likely pleiotropic in neuropsychiatric traits and to harbor human-specific regulatory elements (HSREs). Candidate genes associated with human cortical morphology indeed showed human-specific expression in adult brain cortices, particularly in neuroglia likely regulated by HSREs. Expression levels of candidate genes across human brain developmental stages showed a clear pattern of uplifted expression in early brain development crucial to RD development. Following the new insights and loci pleiotropic in cognitive traits, we identified four novel genes from the GWAS sub-significant associations (i.e., FOXO3, MAPT, KMT2E and HTT) and the Semaphorin gene family with functional priors (i.e., SEMA3A, SEMA3E and SEMA5B). These novel genes were related to neuronal plasticity and disorders, mostly conserved the pattern of uplifted expression in early brain development and had evident expression in cortical neuroglial cells. Our findings jointly illuminated the association of RD with neuroglia regulation-an emerging hotspot in studying neurodevelopmental disorders, and highlighted the need of improving RD phenotyping to avoid jeopardizing future genetic studies of RD.
Collapse
Affiliation(s)
- Wen‐Hua Wei
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary CareThe University of ManchesterManchesterUK
| | - Shaowei Ma
- Hebei Key Laboratory of Children's Cognition and Digital Education and School of Foreign LanguagesLangfang Normal UniversityLangfangChina
| | - Bo Fu
- School of Data ScienceFudan UniversityShanghaiChina
| | - Ranran Song
- Department of Maternal and Child Health and MOE (Ministry of Education) Key Laboratory of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hui Guo
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary CareThe University of ManchesterManchesterUK
| |
Collapse
|
16
|
Li L, Chen Z, Hao C. Neuroprotective effects of polyphyllin VI against rotenone-induced toxicity in SH-SY5Y cells. Brain Res 2024; 1830:148824. [PMID: 38417654 DOI: 10.1016/j.brainres.2024.148824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND A substantial body of evidence is drawing connections between Parkinson's disease (PD) and the phenomena of oxidative stress and mitochondrial dysfunction. Polyphyllin VI (PPVI), an active compound found in Rhizoma Paridis-commonly known as Chonglou (CL) in China, has been identified for its various pharmacological properties, including anti-tumor and anti-inflammatory effects. OBJECTIVE In the present study, an in vitro model of PD was established by treating SH-SY5Y cells with rotenone (ROT), to evaluate the potential neuroprotective effects of polyphyllin VI and its underlying mechanism. METHODS SH-SY5Y cells were treated with ROT to establish an in vitro model of PD. The effects of polyphyllin VI on cell viability were assessed using the resazurin assay. Cell morphology was examined using a microscope. The YO-PRO-1/PI was used to detect apoptosis. Mito-Tracker Red CMXRos, Mito-Tracker Green, and JC-1 were used to detect the effects of polyphyllin Ⅵ on mitochondrial viability, morphology, and function. Oxidative stress-related marker detection kits were used to identify the effects of polyphyllin VI on oxidative stress. Western blot analysis was employed to investigate the signaling pathways associated with neuroprotection. RESULTS PPVI increased ROT-induced SH-SY5Y cell viability and improved ROT-induced cellular morphological changes. PPVI ameliorated ROT-induced oxidative stress status, and attenuated mitochondrial function and morphological changes. PPVI may exert neuroprotective effects through FOXO3α/CREB1/DJ-1-related signaling pathways. CONCLUSION These preliminary findings suggested that PPVI possesses neuroprotective attributes in vitro, and it may be a potential candidate for PD treatment. However, extensive research is necessary to fully understand the mechanisms of PPVI and its effectiveness both in vitro and in vivo.
Collapse
Affiliation(s)
- Lanxin Li
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhengqian Chen
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Cui Hao
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
17
|
Swindell WR. Meta-analysis of differential gene expression in lower motor neurons isolated by laser capture microdissection from post-mortem ALS spinal cords. Front Genet 2024; 15:1385114. [PMID: 38689650 PMCID: PMC11059082 DOI: 10.3389/fgene.2024.1385114] [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: 02/11/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction ALS is a fatal neurodegenerative disease for which underlying mechanisms are incompletely understood. The motor neuron is a central player in ALS pathogenesis but different transcriptome signatures have been derived from bulk analysis of post-mortem tissue and iPSC-derived motor neurons (iPSC-MNs). Methods This study performed a meta-analysis of six gene expression studies (microarray and RNA-seq) in which laser capture microdissection (LCM) was used to isolate lower motor neurons from post-mortem spinal cords of ALS and control (CTL) subjects. Differentially expressed genes (DEGs) with consistent ALS versus CTL expression differences across studies were identified. Results The analysis identified 222 ALS-increased DEGs (FDR <0.10, SMD >0.80) and 278 ALS-decreased DEGs (FDR <0.10, SMD < -0.80). ALS-increased DEGs were linked to PI3K-AKT signaling, innate immunity, inflammation, motor neuron differentiation and extracellular matrix. ALS-decreased DEGs were associated with the ubiquitin-proteosome system, microtubules, axon growth, RNA-binding proteins and synaptic membrane. ALS-decreased DEG mRNAs frequently interacted with RNA-binding proteins (e.g., FUS, HuR). The complete set of DEGs (increased and decreased) overlapped significantly with genes near ALS-associated SNP loci (p < 0.01). Transcription factor target motifs with increased proximity to ALS-increased DEGs were identified, most notably DNA elements predicted to interact with forkhead transcription factors (e.g., FOXP1) and motor neuron and pancreas homeobox 1 (MNX1). Some of these DNA elements overlie ALS-associated SNPs within known enhancers and are predicted to have genotype-dependent MNX1 interactions. DEGs were compared to those identified from SOD1-G93A mice and bulk spinal cord segments or iPSC-MNs from ALS patients. There was good correspondence with transcriptome changes from SOD1-G93A mice (r ≤ 0.408) but most DEGs were not differentially expressed in bulk spinal cords or iPSC-MNs and transcriptome-wide effect size correlations were weak (bulk tissue: r ≤ 0.207, iPSC-MN: r ≤ 0.037). Conclusion This study defines a robust transcriptome signature from LCM-based motor neuron studies of post-mortem tissue from ALS and CTL subjects. This signature differs from those obtained from analysis of bulk spinal cord segments and iPSC-MNs. Results provide insight into mechanisms underlying gene dysregulation in ALS and highlight connections between these mechanisms, ALS genetics, and motor neuron biology.
Collapse
Affiliation(s)
- William R. Swindell
- Department of Internal Medicine, Division of Hospital Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
18
|
Pandaram A, Paul J, Wankhar W, Thakur A, Verma S, Vasudevan K, Wankhar D, Kammala AK, Sharma P, Jaganathan R, Iyaswamy A, Rajan R. Aspartame Causes Developmental Defects and Teratogenicity in Zebra Fish Embryo: Role of Impaired SIRT1/FOXO3a Axis in Neuron Cells. Biomedicines 2024; 12:855. [PMID: 38672209 PMCID: PMC11048232 DOI: 10.3390/biomedicines12040855] [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: 03/07/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Aspartame, a widely used artificial sweetener, is present in many food products and beverages worldwide. It has been linked to potential neurotoxicity and developmental defects. However, its teratogenic effect on embryonic development and the underlying potential mechanisms need to be elucidated. We investigated the concentration- and time-dependent effects of aspartame on zebrafish development and teratogenicity. We focused on the role of sirtuin 1 (SIRT1) and Forkhead-box transcription factor (FOXO), two proteins that play key roles in neurodevelopment. It was found that aspartame exposure reduced the formation of larvae and the development of cartilage in zebrafish. It also delayed post-fertilization development by altering the head length and locomotor behavior of zebrafish. RNA-sequencing-based DEG analysis showed that SIRT1 and FOXO3a are involved in neurodevelopment. In silico and in vitro analyses showed that aspartame could target and reduce the expression of SIRT1 and FOXO3a proteins in neuron cells. Additionally, aspartame triggered the reduction of autophagy flux by inhibiting the nuclear translocation of SIRT1 in neuronal cells. The findings suggest that aspartame can cause developmental defects and teratogenicity in zebrafish embryos and reduce autophagy by impairing the SIRT1/FOXO3a axis in neuron cells.
Collapse
Affiliation(s)
- Athiram Pandaram
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Chennai 600113, Tamil Nadu, India
| | - Jeyakumari Paul
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Chennai 600113, Tamil Nadu, India
| | - Wankupar Wankhar
- Faculty of Paramedical Sciences, Assam down town University, Guwahati 781026, Assam, India
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Sakshi Verma
- Department of Pharmacy, Usha Martin University, Ranchi 835103, Jharkhand, India
| | - Karthick Vasudevan
- Department of Biotechnology, REVA University, Bangalore 560064, Karnataka, India
| | - Dapkupar Wankhar
- Faculty of Paramedical Sciences, Assam down town University, Guwahati 781026, Assam, India
| | - Ananth Kumar Kammala
- Department of Obstetrics and Gynaecology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Priyanshu Sharma
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ravindran Jaganathan
- Preclinical Department, Faculty of Medicine, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh 30450, Perak, Malaysia
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Ravindran Rajan
- Department of Physiology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Chennai 600113, Tamil Nadu, India
| |
Collapse
|
19
|
Jahan S, Ansari UA, Srivastava AK, Aldosari S, Alabdallat NG, Siddiqui AJ, Khan A, Albadrani HM, Sarkar S, Khan B, Adnan M, Pant AB. A protein-miRNA biomic analysis approach to explore neuroprotective potential of nobiletin in human neural progenitor cells (hNPCs). Front Pharmacol 2024; 15:1343569. [PMID: 38348393 PMCID: PMC10860404 DOI: 10.3389/fphar.2024.1343569] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/04/2024] [Indexed: 02/15/2024] Open
Abstract
Chemical-induced neurotoxicity is increasingly recognized to accelerate the development of neurodegenerative disorders (NDs), which pose an increasing health burden to society. Attempts are being made to develop drugs that can cross the blood-brain barrier and have minimal or no side effects. Nobiletin (NOB), a polymethoxylated flavonoid with anti-oxidative and anti-inflammatory effects, has been demonstrated to be a promising compound to treat a variety of NDs. Here, we investigated the potential role of NOB in sodium arsenate (NA)-induced deregulated miRNAs and target proteins in human neural progenitor cells (hNPCs). The proteomics and microRNA (miRNA) profiling was done for different groups, namely, unexposed control, NA-exposed, NA + NOB, and NOB groups. Following the correlation analysis between deregulated miRNAs and target proteins, RT-PCR analysis was used to validate the selected genes. The proteomic analysis showed that significantly deregulated proteins were associated with neurodegeneration pathways, response to oxidative stress, RNA processing, DNA repair, and apoptotic process following exposure to NA. The OpenArray analysis confirmed that NA exposure significantly altered miRNAs that regulate P53 signaling, Wnt signaling, cell death, and cell cycle pathways. The RT-PCR validation studies concur with proteomic data as marker genes associated with autophagy and apoptosis (HO-1, SQSTM1, LC-3, Cas3, Apaf1, HSP70, and SNCA1) were altered following NA exposure. It was observed that the treatment of NOB significantly restored the deregulated miRNAs and proteins to their basal levels. Hence, it may be considered one of its neuroprotective mechanisms. Together, the findings are promising to demonstrate the potential applicability of NOB as a neuroprotectant against chemical-induced neurotoxicity.
Collapse
Affiliation(s)
- Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, 11952, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, 11952 Majmaah, Saudi Arabia
| | - Uzair Ahmad Ansari
- Developmental Toxicology Laboratory, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ankur Kumar Srivastava
- Developmental Toxicology Laboratory, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow 226001, Uttar Pradesh, India
| | - Sahar Aldosari
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, 11952, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, 11952 Majmaah, Saudi Arabia
| | - Nessrin Ghazi Alabdallat
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, 11952, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, 11952 Majmaah, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, Uttar Pradesh 226026, India
| | - Hind Muteb Albadrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province 34212, Saudi Arabia
| | - Sana Sarkar
- Developmental Toxicology Laboratory, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow 226001, Uttar Pradesh, India
| | - Bushra Khan
- Developmental Toxicology Laboratory, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow 226001, Uttar Pradesh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Aditya Bhushan Pant
- Developmental Toxicology Laboratory, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow 226001, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
20
|
Narasimhamurthy RK, Venkidesh BS, Nayak S, Reghunathan D, Mallya S, Sharan K, Rao BSS, Mumbrekar KD. Low-dose exposure to malathion and radiation results in the dysregulation of multiple neuronal processes, inducing neurotoxicity and neurodegeneration in mouse. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:1403-1418. [PMID: 38038914 PMCID: PMC10789675 DOI: 10.1007/s11356-023-31085-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023]
Abstract
Neurodegenerative disorders are a debilitating and persistent threat to the global elderly population, carrying grim outcomes. Their genesis is often multifactorial, with a history of prior exposure to xenobiotics such as pesticides, heavy metals, enviornmental pollutants, ionizing radiation etc,. A holistic molecular insight into their mechanistic induction upon single or combinatorial exposure to different toxicants is still unclear. In the present study, one-month-old C57BL/6 male mice were administered orally with malathion (50 mg/kg body wt. for 14 days) and single whole-body radiation (0.5 Gy) on the 8th day. Post-treatment, behavioural assays for exploratory behaviour, memory, and learning were performed. After sacrifice, brains were collected for histology, biochemical assays, and transcriptomic analysis. Transcriptomic analysis revealed several altered processes like synaptic transmission and plasticity, neuronal survival, proliferation, and death. Signalling pathways like MAPK, PI3K-Akt, Apelin, NF-κB, cAMP, Notch etc., and pathways related to neurodegenerative diseases were altered. Increased astrogliosis was observed in the radiation and coexposure groups, with significant neuronal cell death and a reduction in the expression of NeuN. Sholl analysis, dendritic arborization and spine density studies revealed decreased total apical neuronal path length and dendritic spine density. Reduced levels of the antioxidants GST and GSH and acetylcholinesterase enzyme activity were also detected. However, no changes were seen in exploratory behaviour or learning and memory post-treatment. Thus, explicating the molecular mechanisms behind malathion and radiation can provide novel insights into external factor-driven neurotoxicity and neurodegenerative pathogenesis.
Collapse
Affiliation(s)
- Rekha Koravadi Narasimhamurthy
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Babu Santhi Venkidesh
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sangeetha Nayak
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Dinesh Reghunathan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Krishna Sharan
- Department of Radiotherapy, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Bola Sadashiva Satish Rao
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Directorate of Research, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Kamalesh Dattaram Mumbrekar
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
21
|
Chen L, Xu W, Zhang Y, Chen H, Han Y. Gandouling alleviates nerve injury through PI3K/Akt/FoxO1 and Sirt1/FoxO1 signaling pathway to inhibit autophagy in the rats model of Wilson's disease. Brain Behav 2023; 13:e3325. [PMID: 38010098 PMCID: PMC10726812 DOI: 10.1002/brb3.3325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/18/2023] [Accepted: 10/18/2023] [Indexed: 11/29/2023] Open
Abstract
INTRODUCTION Previous studies have shown that Gandouling (GDL) may alleviate the nerve damage caused by Wilson's disease (WD) by inhibiting the autophagy of nerve cell mitochondria. However, its mechanisms are still unclear. Revealing the therapeutic mechanism of GDL is beneficial for its clinical application and provides theoretical support for the development of new formulations for treating WD. METHOD This time we found that the oxidative stress level in the body of the copper-overloaded WD rates increased, neurons in the hippocampus were damaged, and autophagy occurred. GDL reversed these situations and significantly improved the learning, memory, and spatial cognitive abilities of the high-copper-loaded WD rates. After GDL intervention, the expression of phosphatidylinositol-3 kinase (PI3K), phosphorylated serine-threonine protein kinase (AKT), and phosphorylated forkhead box protein O1 (FoxO1) significantly increased, whereas FoxO1 in the nucleus decreased and phosphorylated FoxO1 in the cytoplasm also significantly raised. In addition, the expression of Sirt1 significantly declined, and Ac-FoxO1 in the nucleus also significantly increased. RESULTS These data indicated that GDL may promote the phosphorylation of FoxO1 and promote its nucleation by activating the PI3K/AKT/FoxO1 signaling pathway and inhibit Ac-FoxO1 hydrolysis in the nucleus through the Sirt1/FoxO1 signaling pathway to suppress the transcriptional activity of FoxO1. CONCLUSION Furthermore, it inhibited the expression of autophagy genes Atg12 and Gabarapl1. In summary, our work provides new insights into the potential mechanisms of GDL repairing WD neuronal damage through autophagy pathways.
Collapse
Affiliation(s)
- Li Chen
- Institute of Pharmaceutical DepartmentThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiP. R. China
| | - Wangyang Xu
- Institute of school of pharmacyAnhui University of Chinese MedicineHefeiP. R. China
| | - Yuting Zhang
- Institute of Pharmaceutical DepartmentThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiP. R. China
| | - Hao Chen
- Institute of Pharmaceutical DepartmentThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiP. R. China
| | - Yanquan Han
- Institute of Pharmaceutical DepartmentThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiP. R. China
| |
Collapse
|
22
|
Donlon TA, Morris BJ, Chen R, Lim E, Morgen EK, Fortney K, Shah N, Masaki KH, Willcox BJ. Proteomic basis of mortality resilience mediated by FOXO3 longevity genotype. GeroScience 2023; 45:2303-2324. [PMID: 36881352 PMCID: PMC10651822 DOI: 10.1007/s11357-023-00740-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/23/2023] [Indexed: 03/08/2023] Open
Abstract
FOXO3 is a ubiquitous transcription factor expressed in response to cellular stress caused by nutrient deprivation, inflammatory cytokines, reactive oxygen species, radiation, hypoxia, and other factors. We showed previously that the association of inherited FOXO3 variants with longevity was the result of partial protection against mortality risk posed by aging-related life-long stressors, particularly cardiometabolic disease. We then referred to the longevity-associated genotypes as conferring "mortality resilience." Serum proteins whose levels change with aging and are associated with mortality risk may be considered as "stress proteins." They may serve as indirect measures of life-long stress. Our aims were to (1) identify stress proteins that increase with aging and are associated with an increased risk of mortality, and (2) to determine if FOXO3 longevity/resilience genotype dampens the expected increase in mortality risk they pose. A total of 4500 serum protein aptamers were quantified using the Somalogic SomaScan proteomics platform in the current study of 975 men aged 71-83 years. Stress proteins associated with mortality were identified. We then used age-adjusted multivariable Cox models to investigate the interaction of stress protein with FOXO3 longevity-associated rs12212067 genotypes. For all the analyses, the p values were corrected for multiple comparisons by false discovery rate. This led to the identification of 44 stress proteins influencing the association of FOXO3 genotype with reduced mortality. Biological pathways were identified for these proteins. Our results suggest that the FOXO3 resilience genotype functions by reducing mortality in pathways related to innate immunity, bone morphogenetic protein signaling, leukocyte migration, and growth factor response.
Collapse
Affiliation(s)
- Timothy A Donlon
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Brian J Morris
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA.
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA.
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia.
| | - Randi Chen
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
| | - Eunjung Lim
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Eric K Morgen
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Kristen Fortney
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Naisha Shah
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Kamal H Masaki
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Bradley J Willcox
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
23
|
Fredi BM, De Labio RW, Rasmussen LT, Chagas EFB, Chen ES, Turecki G, Smith MDAC, Payão SLM. CDK10, CDK11, FOXO1, and FOXO3 Gene Expression in Alzheimer's Disease Encephalic Samples. Cell Mol Neurobiol 2023; 43:2953-2962. [PMID: 36988771 PMCID: PMC11410123 DOI: 10.1007/s10571-023-01341-9] [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: 11/27/2022] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD) is a progressive neuroinflammatory and neurodegenerative disorder that affects different regions of the brain. Its pathophysiology includes the accumulation of β-amyloid protein, formation of neurofibrillary tangles, and inflammatory processes. Genetic factors are involved in the onset of AD, but they are not fully elucidated. Identification of gene expression in encephalic tissues of patients with AD may help elucidate its development. Our objectives were to characterize and compare the gene expression of CDK10, CDK11, FOXO1, and FOXO3 in encephalic tissue samples from AD patients and elderly controls, from the auditory cortex and cerebellum. RT-qPCR was used on samples from 82 individuals (45 with AD and 37 controls). We observed a statistically significant increase in CDK10 (p = 0.029*) and CDK11 (p = 0.048*) gene expression in the AD group compared to the control, which was most evident in the cerebellum. Furthermore, the Spearman test demonstrated the presence of a positive correlation of gene expression both in the auditory cortex in the AD group (r = 0.046/p = 0.004) and control group (r = 0.454/p = 0.005); and in the cerebellum in the AD group (r = 0.654 /p < 0.001). There was no statistically significant difference and correlation in the gene expression of FOXO1 and FOXO3 in the AD group and the control. In conclusion, CDK10 and CDK11 have high expression in AD patients compared to control, and they present a positive correlation of gene expression in the analyzed groups and tissues, which suggests that they play an important role in the pathogenesis of AD.
Collapse
Affiliation(s)
| | | | | | | | - Elizabeth Suchi Chen
- Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Gustavo Turecki
- The Douglas-Bell Canada Brain Bank, Douglas Mental Health University, Montreal, QC, Canada
| | | | | |
Collapse
|
24
|
Liu Y, Liu Q, Zhang Z, Yang Y, Zhou Y, Yan H, Wang X, Li X, Zhao J, Hu J, Yang S, Tian Y, Yao Y, Qiu Z, Song Y, Yang Y. The regulatory role of PI3K in ageing-related diseases. Ageing Res Rev 2023; 88:101963. [PMID: 37245633 DOI: 10.1016/j.arr.2023.101963] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 05/30/2023]
Abstract
Ageing is a physiological/pathological process accompanied by the progressive damage of cell function, triggering various ageing-related disorders. Phosphatidylinositol 3-kinase (PI3K), which serves as one of the central regulators of ageing, is closely associated with cellular characteristics or molecular features, such as genome instability, telomere erosion, epigenetic alterations, and mitochondrial dysfunction. In this review, the PI3K signalling pathway was firstly thoroughly explained. The link between ageing pathogenesis and the PI3K signalling pathway was then summarized. Finally, the key regulatory roles of PI3K in ageing-related illnesses were investigated and stressed. In summary, we revealed that drug development and clinical application targeting PI3K is one of the focal points for delaying ageing and treating ageing-related diseases in the future.
Collapse
Affiliation(s)
- Yanqing Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Zhe Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yaru Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yazhe Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Huanle Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Xin Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Xiaoru Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Jing Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Jingyan Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Shulin Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yifan Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yu Yao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Zhenye Qiu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yanbin Song
- Department of Cardiology, Affiliated Hospital, Yan'an University, 43 North Street, Yan'an 716000, China.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| |
Collapse
|
25
|
Wang M, Yu H, He Y, Liao S, Xu D. Cross-talk between traditional Chinese medicine and Parkinson's disease based on cell autophagy. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 7:100235. [DOI: 10.1016/j.prmcm.2023.100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
26
|
Zhao T, Miao H, Song Z, Li Y, Xia N, Zhang Z, Zhang H. Metformin alleviates the cognitive impairment induced by benzo[a]pyrene via glucolipid metabolism regulated by FTO/FoxO6 pathway in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:69192-69204. [PMID: 37133670 DOI: 10.1007/s11356-023-27303-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/25/2023] [Indexed: 05/04/2023]
Abstract
Benzo[a]pyrene (B[a]P) is neurotoxic; however, the mechanism and prevention are still unclear. In this study, we assessed the intervention effect of metformin (MET) on cognitive dysfunction in mice induced by B[a]P from the perspective of glucolipid metabolism. Forty-two male healthy ICR mice were randomly categorized into 6 groups and were gavaged with B[a]P (0, 2.5, 5, or 10 mg/kg), 45 times for 90 days. The controls were gavaged with edible peanut oil, and the intervention groups were co-treated with B[a]P (10 mg/kg) and MET (200 or 300 mg/kg). We assessed the cognitive function of mice, observed the pathomorphological and ultrastructural changes, and detected neuronal apoptosis and glucolipid metabolism. Results showed that B[a]P dose-dependently induced cognitive impairment, neuronal damage, glucolipid metabolism disorder in mice, and enhanced proteins of fat mass and obesity-associated protein (FTO) and forkhead box protein O6 (FoxO6) in the cerebral cortex and liver, which were alleviated by the MET intervention. The findings indicated the critical role of glucolipid metabolism disorder in the cognitive impairment in mice caused by B[a]P and the prevention of MET against B[a]P neurotoxicity by regulating glucolipid metabolism via restraining FTO/FoxO6 pathway. The finding provides a scientific basis for the neurotoxicity and prevention strategies of B[a]P.
Collapse
Affiliation(s)
- Tingyi Zhao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China
| | - Huide Miao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China
| | - Zhanfei Song
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China
| | - Yangyang Li
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China
| | - Na Xia
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China
| | - Zhiyan Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan, China.
| |
Collapse
|
27
|
Yu J, Li T, Zhu J. Gene Therapy Strategies Targeting Aging-Related Diseases. Aging Dis 2023; 14:398-417. [PMID: 37008065 PMCID: PMC10017145 DOI: 10.14336/ad.2022.00725] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Rapid advancements have taken place in gene therapy technology. However, effective methods for treating aging- or age-related chronic diseases, which are often closely related to genes or even multiple genes, are still lacking. The path to developing cures is winding, while gene therapy that targets genes related to aging represents an exciting research direction with tremendous potential. Among aging-related genes, some candidates have been studied at different levels, from cell to organismal levels (e.g., mammalian models) with different methods, from overexpression to gene editing. The TERT and APOE have even entered the stage of clinical trials. Even those displaying only a preliminary association with diseases have potential applications. This article discusses the foundations and recent breakthroughs in the field of gene therapy, providing a summary of current mainstream strategies and gene therapy products with clinical and preclinical applications. Finally, we review representative target genes and their potential for treating aging or age-related diseases.
Collapse
Affiliation(s)
| | | | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, National Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai, China.
| |
Collapse
|
28
|
Redenšek Trampuž S, Vogrinc D, Goričar K, Dolžan V. Shared miRNA landscapes of COVID-19 and neurodegeneration confirm neuroinflammation as an important overlapping feature. Front Mol Neurosci 2023; 16:1123955. [PMID: 37008787 PMCID: PMC10064073 DOI: 10.3389/fnmol.2023.1123955] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction Development and worsening of most common neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, have been associated with COVID-19 However, the mechanisms associated with neurological symptoms in COVID-19 patients and neurodegenerative sequelae are not clear. The interplay between gene expression and metabolite production in CNS is driven by miRNAs. These small non-coding molecules are dysregulated in most common neurodegenerative diseases and COVID-19. Methods We have performed a thorough literature screening and database mining to search for shared miRNA landscapes of SARS-CoV-2 infection and neurodegeneration. Differentially expressed miRNAs in COVID-19 patients were searched using PubMed, while differentially expressed miRNAs in patients with five most common neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis) were searched using the Human microRNA Disease Database. Target genes of the overlapping miRNAs, identified with the miRTarBase, were used for the pathway enrichment analysis performed with Kyoto Encyclopedia of Genes and Genomes and Reactome. Results In total, 98 common miRNAs were found. Additionally, two of them (hsa-miR-34a and hsa-miR-132) were highlighted as promising biomarkers of neurodegeneration, as they are dysregulated in all five most common neurodegenerative diseases and COVID-19. Additionally, hsa-miR-155 was upregulated in four COVID-19 studies and found to be dysregulated in neurodegeneration processes as well. Screening for miRNA targets identified 746 unique genes with strong evidence for interaction. Target enrichment analysis highlighted most significant KEGG and Reactome pathways being involved in signaling, cancer, transcription and infection. However, the more specific identified pathways confirmed neuroinflammation as being the most important shared feature. Discussion Our pathway based approach has identified overlapping miRNAs in COVID-19 and neurodegenerative diseases that may have a valuable potential for neurodegeneration prediction in COVID-19 patients. Additionally, identified miRNAs can be further explored as potential drug targets or agents to modify signaling in shared pathways. Graphical AbstractShared miRNA molecules among the five investigated neurodegenerative diseases and COVID-19 were identified. The two overlapping miRNAs, hsa-miR-34a and has-miR-132, present potential biomarkers of neurodegenerative sequelae after COVID-19. Furthermore, 98 common miRNAs between all five neurodegenerative diseases together and COVID-19 were identified. A KEGG and Reactome pathway enrichment analyses was performed on the list of shared miRNA target genes and finally top 20 pathways were evaluated for their potential for identification of new drug targets. A common feature of identified overlapping miRNAs and pathways is neuroinflammation. AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; COVID-19, coronavirus disease 2019; HD, Huntington's disease; KEGG, Kyoto Encyclopedia of Genes and Genomes; MS, multiple sclerosis; PD, Parkinson's disease.
Collapse
Affiliation(s)
| | | | | | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
29
|
Yan B, Jin Y, Mao S, Zhang Y, Yang D, Du M, Yin Y. Smurf2-Mediated Ubiquitination of FOXO4 Regulates Oxygen-glucose Deprivation/Reperfusion-induced Pyroptosis of Cortical Neurons. Curr Neurovasc Res 2023; 20:443-452. [PMID: 37861000 DOI: 10.2174/0115672026267629230920062917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Smad ubiquitination regulatory factor 2 (Smurf2) has been observed to alleviate ischemia-reperfusion injury. This study sought to explore the molecular mechanism of Smurf2-mediated forkhead box O4 (FOXO4) ubiquitination in oxygen-glucose deprivation/ reperfusion (OGD/R)-induced pyroptosis of cortical neurons. METHODS Human cortical neurons (HCN-2) were subjected to OGD/R to establish a cell model of cerebral stroke. Smurf2, FOXO4, and doublecortin domain containing 2 (DCDC2) expressions were determined by RT-qPCR and Western blot. LDH release, pyroptosis-related proteins NLRP3, GSDMD-N, and cleaved-caspase-3, as well as inflammatory factors IL-1β and IL-18, were assessed by LDH assay kit, Western blot, and ELISA. The ubiquitination level of FOXO4 was determined by ubiquitination assay. The bindings of Smurf2 to FOXO4 and FOXO4 to DCDC2 were testified by Co-IP, ChIP, and dual-luciferase assays. Rescue experiments were designed to validate the role of FOXO4/DCDC2 in the pyroptosis of HCN-2 cells. RESULTS Smurf2 was weakly expressed, while FOXO4 and DCDC2 were prominently expressed in OGD/R-treated HCN-2 cells. Smurf2 overexpression promoted LDH release, reduced NLRP3, GSDMD-N, and cleaved-caspase-3 proteins, and decreased IL-1β and IL-18 concentrations. Sumrf2 improved the ubiquitination level of FOXO4 to downregulate its protein level. FOXO4 is bound to the DCDC2 promoter to facilitate its transcription. Overexpression of FOXO4 or DCDC2 reversed the inhibition of Smurf2 overexpression on pyroptosis of OGD/Rtreated HCN-2 cells. CONCLUSION Smurf2 overexpression facilitated the ubiquitination of FOXO4 to reduce its protein level, thereby suppressing DCDC2 transcription and restricting OGD/R-induced pyroptosis of cortical neurons.
Collapse
Affiliation(s)
- Bin Yan
- Department of Geriatric Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Jin
- Department of Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Song Mao
- Department of Pediatrics, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhang
- Department of Neurology, Chenzhou First People's Hospital, Chenzhou, China
| | - Dahong Yang
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Mingyang Du
- Cerebrovascular Disease Treatment Center, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yugang Yin
- Department of Geriatric Cardiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
30
|
Wang Y, Lee H, Fear JM, Berger I, Oliver B, Przytycka TM. NetREX-CF integrates incomplete transcription factor data with gene expression to reconstruct gene regulatory networks. Commun Biol 2022; 5:1282. [PMID: 36418514 PMCID: PMC9684490 DOI: 10.1038/s42003-022-04226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
The inference of Gene Regulatory Networks (GRNs) is one of the key challenges in systems biology. Leading algorithms utilize, in addition to gene expression, prior knowledge such as Transcription Factor (TF) DNA binding motifs or results of TF binding experiments. However, such prior knowledge is typically incomplete, therefore, integrating it with gene expression to infer GRNs remains difficult. To address this challenge, we introduce NetREX-CF-Regulatory Network Reconstruction using EXpression and Collaborative Filtering-a GRN reconstruction approach that brings together Collaborative Filtering to address the incompleteness of the prior knowledge and a biologically justified model of gene expression (sparse Network Component Analysis based model). We validated the NetREX-CF using Yeast data and then used it to construct the GRN for Drosophila Schneider 2 (S2) cells. To corroborate the GRN, we performed a large-scale RNA-Seq analysis followed by a high-throughput RNAi treatment against all 465 expressed TFs in the cell line. Our knockdown result has not only extensively validated the GRN we built, but also provides a benchmark that our community can use for evaluating GRNs. Finally, we demonstrate that NetREX-CF can infer GRNs using single-cell RNA-Seq, and outperforms other methods, by using previously published human data.
Collapse
Affiliation(s)
- Yijie Wang
- Computer Science Department, Indiana University, Bloomington, IN, 47408, USA.
| | - Hangnoh Lee
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA
| | - Justin M Fear
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA
| | - Isabelle Berger
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA
| | - Brian Oliver
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA.
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD, 20894, USA.
| |
Collapse
|
31
|
Therapeutic Effect of Melatonin in Premature Ovarian Insufficiency: Hippo Pathway Is Involved. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3425877. [PMID: 36017238 PMCID: PMC9398856 DOI: 10.1155/2022/3425877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
Abstract
Objective Premature ovarian insufficiency (POI) is a female reproductive disorder of unknown etiology with no definite pathogenesis. Melatonin (MT) is an endogenous hormone synthesized mainly by pineal cells and has strong endogenous effects in regulating ovarian function. To systematically explore the pharmacological mechanism of MT on POI therapy, a literature review approach was conducted at the signaling pathways level. Methods Relevant literatures were searched and downloaded from databases, including PubMed and China National Knowledge Infrastructure, using the keywords “premature ovarian insufficiency,” “Hippo signaling pathways,” and “melatonin.” The search criteria were from 2010 to 2022. Text mining was also performed. Results MT is involved in the regulation of Hippo signaling pathway in a variety of modes and has been correlated with ovarian function. Conclusions The purpose of this review is to summarize the research progress of Hippo signaling pathways and significance of MT in POI, the potential crosstalk between MT and Hippo signaling pathways, and the prospective therapy.
Collapse
|
32
|
Zhou Z, Zhou J, Liao J, Chen Z, Zheng Y. The Emerging Role of Astrocytic Autophagy in Central Nervous System Disorders. Neurochem Res 2022; 47:3697-3708. [PMID: 35960484 DOI: 10.1007/s11064-022-03714-w] [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: 03/13/2022] [Revised: 07/15/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Astrocytes act as "housekeeping cells" for maintaining cerebral homeostasis and play an important role in many disorders. Recent studies further highlight the contribution of autophagy to astrocytic functions, including astrogenesis, the astrocytic removal of neurotoxins or stressors, and astrocytic polarization. More importantly, genetic and pharmacological approaches have provided evidence that outlines the contributions of astrocytic autophagy to several brain disorders, including neurodegeneration, cerebral ischemia, and depression. In this study, we summarize the emerging role of autophagy in regulating astrocytic functions and discuss the contributions of astrocytic autophagy to different CNS disorders.
Collapse
Affiliation(s)
- Zhuchen Zhou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Zhou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jie Liao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
33
|
Wiegreffe C, Wahl T, Joos NS, Bonnefont J, Liu P, Britsch S. Developmental cell death of cortical projection neurons is controlled by a Bcl11a/Bcl6‐dependent pathway. EMBO Rep 2022; 23:e54104. [PMID: 35766181 PMCID: PMC9346488 DOI: 10.15252/embr.202154104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 12/05/2022] Open
Abstract
Developmental neuron death plays a pivotal role in refining organization and wiring during neocortex formation. Aberrant regulation of this process results in neurodevelopmental disorders including impaired learning and memory. Underlying molecular pathways are incompletely determined. Loss of Bcl11a in cortical projection neurons induces pronounced cell death in upper‐layer cortical projection neurons during postnatal corticogenesis. We use this genetic model to explore genetic mechanisms by which developmental neuron death is controlled. Unexpectedly, we find Bcl6, previously shown to be involved in the transition of cortical neurons from progenitor to postmitotic differentiation state to provide a major checkpoint regulating neuron survival during late cortical development. We show that Bcl11a is a direct transcriptional regulator of Bcl6. Deletion of Bcl6 exerts death of cortical projection neurons. In turn, reintroduction of Bcl6 into Bcl11a mutants prevents induction of cell death in these neurons. Together, our data identify a novel Bcl11a/Bcl6‐dependent molecular pathway in regulation of developmental cell death during corticogenesis.
Collapse
Affiliation(s)
| | - Tobias Wahl
- Institute of Molecular and Cellular Anatomy Ulm University Ulm Germany
| | | | - Jerome Bonnefont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI) Université Libre de Bruxelles (ULB) Brussels Belgium
- VIB‐KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neuroscience Leuven Brain Institute Leuven Belgium
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Stefan Britsch
- Institute of Molecular and Cellular Anatomy Ulm University Ulm Germany
| |
Collapse
|
34
|
Sanchez-Mirasierra I, Ghimire S, Hernandez-Diaz S, Soukup SF. Targeting Macroautophagy as a Therapeutic Opportunity to Treat Parkinson's Disease. Front Cell Dev Biol 2022; 10:921314. [PMID: 35874822 PMCID: PMC9298504 DOI: 10.3389/fcell.2022.921314] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Macroautophagy, an evolutionary conserved catabolic process in the eukaryotic cell, regulates cellular homeostasis and plays a decisive role in self-engulfing proteins, protein aggregates, dysfunctional or damaged organelles, and invading pathogens. Growing evidence from in vivo and in vitro models shows that autophagy dysfunction plays decisive role in the pathogenesis of various neurodegenerative diseases, including Parkinson's disease (PD). PD is an incurable and second most common neurodegenerative disease characterised by neurological and motor dysfunction accompanied of non-motor symptoms that can also reduce the life quality of patients. Despite the investment in research, the aetiology of the disease is still unknown and the therapies available are aimed mostly at ameliorating motor symptoms. Hence, therapeutics regulating the autophagy pathway might play an important role controlling the disease progression, reducing neuronal loss and even ameliorating non-motor symptoms. In this review, we highlight potential therapeutic opportunities involved in different targeting options like an initiation of autophagy, Leucine-rich repeat kinase 2 (LRRK2) inhibition, mitophagy, lysosomes, lipid metabolism, immune system, gene expression, biomarkers, and also non-pharmacological interventions. Thus, strategies to identify therapeutics targeting the pathways modulating autophagy might hold a future for therapy development against PD.
Collapse
Affiliation(s)
| | - Saurav Ghimire
- Universite Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | | | | |
Collapse
|
35
|
Ramaswamy P, Christopher R, Kumar Pal P, Debnath M, Yadav R. Plasma microRNAs as a Potential Biomarker for Identification of Progressive Supranuclear Palsy. Diagnostics (Basel) 2022; 12:diagnostics12051204. [PMID: 35626359 PMCID: PMC9139891 DOI: 10.3390/diagnostics12051204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 02/04/2023] Open
Abstract
Progressive supranuclear palsy (PSP) is the second most common Parkinsonian disorder with complex etiology. The underlying molecular mechanism of PSP pathogenesis remains unclear. The present study aims to find the feasibility of using plasma miRNAs as novel biomarkers. Plasma-focused qPCR panels were used for microRNA profiling and identified differentially expressed microRNAs in PSP compared to controls. The DIANA-miRPath v3.0 was used to perform KEGG pathway analysis. We then confirmed the expression of selected candidates by RT-qPCR and their clinical utility was assessed by ROC analysis. Profiling data revealed 28 differentially expressed microRNAs in PSP. Five overexpressed miRNAs were selected for further analysis. The KEGG pathway analysis revealed 48 high-risk pathways. The study revealed that as a single marker—miR-19b-3p, miR-33a-5p, miR-130b-3p, miR-136-3p, and miR-210-3p had a specificity of 64.71%, 82.35%, 68.75%, 82.35%, and 70.59% at sensitivity 77.78%, 77.78%, 66.67%, 73.33%, and 66.67%, respectively. The result suggests that circulating plasma miRNAs were altered in PSP compared to control. The findings of this study may provide potential biomarkers and pathways associated with PSP. Further large-scale validation studies are required to confirm the same.
Collapse
Affiliation(s)
- Palaniswamy Ramaswamy
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India; (P.R.); (P.K.P.)
| | - Rita Christopher
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India;
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India; (P.R.); (P.K.P.)
| | - Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India;
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560029, India; (P.R.); (P.K.P.)
- Correspondence:
| |
Collapse
|
36
|
Xie Y, Li Z, Xu H, Ma J, Li T, Shi C, Jin J. Downregulation of Sp1 Inhibits the Expression of HDAC1/SOX10 to Alleviate Neuropathic Pain-like Behaviors after Spinal Nerve Ligation in Mice. ACS Chem Neurosci 2022; 13:1446-1455. [PMID: 35420781 DOI: 10.1021/acschemneuro.2c00091] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Specific protein 1 (Sp1) is a member of the Sp/Kruppel-like factor family, which regulates cellular processes of neurons in the nervous system. This study was performed to examine the regulatory role and the underlying mechanism of transcription factor Sp1 in neuropathic pain (NP)-like behaviors after spinal nerve ligation (SNL). Sp1 and histone deacetylase 1(HDAC1) expressions were determined in the C57BL6 mouse model with NP-like behaviors after SNL, which demonstrated that Sp1 and HDAC1 elevation occurred in neurons in the spinal dorsal horn of SNL mice. The chromatin immunoprecipitation assay verified that Sp1 was bound to the HDAC1 promoter region and HDAC1 to the SRY-box-containing gene 10 (SOX10) promoter region in the spinal dorsal horn. Immunofluorescence was performed to determine Sp1, HDAC1, and SOX10 in the spinal dorsal horn neurons as well as the neuronal marker (NeuN), microglial marker (Iba-1), and astrocyte marker (GFAP). The nociceptive test was performed to characterize the hindlimb paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) of mice 0-10 days after model establishment. Loss- and gain-of-function assays revealed that Sp1 promoted HDAC1 expression, and HDAC1 in turn promoted SOX10 expression. HDAC1 elevation reversed the effects of Sp1 silencing, and the increased PWT and PWL of SNL mice were negated after SOX10 overexpression. Meanwhile, SOX10 also restored the results by Sp1 knockdown. Collectively, downregulating Sp1 alleviates NP-like behaviors after SNL via the HDAC1/SOX10 axis.
Collapse
Affiliation(s)
- Yonggang Xie
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P. R. China
| | - Zhen Li
- Department of Otorhinolaryngology, Yantaishan Hospital, Yantai 264000, P. R. China
| | - Hongyu Xu
- Department of Anesthesiology, Central Hospital of Zibo City, Zibo 255000, P. R. China
| | - Jiahai Ma
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P. R. China
| | - Tao Li
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P. R. China
| | - Cunxian Shi
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P. R. China
| | - Jin Jin
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, P. R. China
| |
Collapse
|
37
|
Wang Q, Li Y, Tan H, Wang Y. Sevoflurane-Induced Apoptosis in the Mouse Cerebral Cortex Follows Similar Characteristics of Physiological Apoptosis. Front Mol Neurosci 2022; 15:873658. [PMID: 35465098 PMCID: PMC9024292 DOI: 10.3389/fnmol.2022.873658] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
General anesthetics are capable of inducing neuronal apoptosis during the rapid synaptogenesis of immature mammalian brains. In this vulnerable time window, physiological apoptosis also occurs to eliminate excess and inappropriately integrated neurons. We previously showed that physiological and ketamine-induced apoptosis in mouse primary somatosensory cortex (S1) followed similar developmental patterns. However, since sevoflurane is more widely used in pediatric anesthesia, and targets mainly on different receptors, as compared with ketamine, it is important to determine whether sevoflurane-induced apoptosis also follows similar developmental patterns as physiological apoptosis or not. Mice at postnatal days 5 (P5) and P9 were anesthetized with 1.5% sevoflurane for 4 h, and the apoptotic neurons in S1 were quantitated by immunohistochemistry. The results showed that sevoflurane raised the levels of apoptosis in S1 without interfering with the developmental patterns of physiological apoptosis. The cells more vulnerable to both physiological and sevoflurane-induced apoptosis shifted from layer V pyramidal neurons at P5 to layers II–IV GABAergic neurons by P9. The magnitude of both sevoflurane-induced and physiological apoptosis was more attenuated at P9 than P5. To determine whether the Akt-FoxO1-PUMA pathway contributes to the developmental decrease in magnitude of both physiological and sevoflurane-induced apoptosis, Western blot was used to measure the levels of related proteins in S1 of P5 and P9 mice. We observed higher levels of antiapoptotic phosphorylated Akt (p-Akt) and phosphorylated FoxO1 (p-FoxO1), and lower levels of the downstream proapoptotic factor PUMA in control and anesthetized mice at P9 than P5. In addition, the Akt-FoxO1-PUMA pathway may also be responsible for sevoflurane-induced apoptosis. Together, these results suggest that magnitude, lamination pattern and cell-type specificity to sevoflurane-induced apoptosis are age-dependent and follow physiological apoptosis pattern. Moreover, The Akt-FoxO1-PUMA pathway may mediate the developmental decreases in magnitude of both physiological and sevoflurane-induced apoptosis in neonatal mouse S1.
Collapse
|
38
|
Guo S, Mangal R, Dandu C, Geng X, Ding Y. Role of Forkhead Box Protein O1 (FoxO1) in Stroke: A Literature Review. Aging Dis 2022; 13:521-533. [PMID: 35371601 PMCID: PMC8947839 DOI: 10.14336/ad.2021.0826] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the most prevalent causes of death around the world. When a stroke occurs, many cellular signaling cascades and regulators are activated, which results in severe cellular dysfunction and debilitating long-term disability. One crucial regulator of cell fate and function is mammalian Forkhead box protein O1 (FoxO1). Many studies have found FoxO1 to be implicated in many cellular processes, including regulating gluconeogenesis and glycogenolysis. During a stroke, modifications of FoxO1 have been linked to a variety of functions, such as inducing cell death and inflammation, inhibiting oxidative injury, affecting the blood brain barrier (BBB), and regulating hepatic gluconeogenesis. For these functions of FoxO1, different measures and treatments were applied to FoxO1 after ischemia. However, the subtle mechanisms of post-transcriptional modification and the role of FoxO1 are still elusive and even contradictory in the development of stroke. The determination of these mechanisms will lead to further enlightenment for FoxO1 signal transduction and the identification of targeted drugs. The regulation and function of FoxO1 may provide an important way for the prevention and treatment of diseases. Overall, the functions of FoxO1 are multifactorial, and this paper will summarize all of the significant pathways in which FoxO1 plays an important role during stroke damage and recovery.
Collapse
Affiliation(s)
- Sichao Guo
- 1Luhe Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ruchi Mangal
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Chaitu Dandu
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaokun Geng
- 1Luhe Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,2Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yuchuan Ding
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
39
|
Li F, Gao J, Kohls W, Geng X, Ding Y. Perspectives on benefit of early and prereperfusion hypothermia by pharmacological approach in stroke. Brain Circ 2022; 8:69-75. [PMID: 35909706 PMCID: PMC9336590 DOI: 10.4103/bc.bc_27_22] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Stroke kills or disables approximately 15 million people worldwide each year. It is the leading cause of brain injury, resulting in persistent neurological deficits and profound physical handicaps. In spite of over 100 clinical trials, stroke treatment modalities are limited in applicability and efficacy, and therefore, identification of new therapeutic modalities is required to combat this growing problem. Poststroke oxidative damage and lactic acidosis are widely-recognized forms of brain ischemia/reperfusion injury. However, treatments directed at these injury mechanisms have not been effective. In this review, we offer a novel approach combining these well-established damage mechanisms with new insights into brain glucose handling. Specifically, emerging evidence of brain gluconeogenesis provides a missing link for understanding oxidative injury and lactate toxicity after ischemia. Therefore, dysfunctional gluconeogenesis may substantially contribute to oxidative and lactate damage. We further review that hypothermia initiated early in ischemia and before reperfusion may ameliorate gluconeogenic dysfunction and subsequently provide an important mechanism of hypothermic protection. We will focus on the efficacy of pharmacologically assisted hypothermia and suggest a combination that minimizes side effects. Together, this study will advance our knowledge of basic mechanisms of ischemic damage and apply this knowledge to develop new therapeutic strategies that are desperately needed in the clinical treatment of stroke.
Collapse
Affiliation(s)
- Fengwu Li
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Jie Gao
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Wesley Kohls
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Neurology, China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
40
|
Liu Y, Xu Y, Yu M. MicroRNA-4722-5p and microRNA-615-3p serve as potential biomarkers for Alzheimer's disease. Exp Ther Med 2022; 23:241. [PMID: 35222718 PMCID: PMC8815048 DOI: 10.3892/etm.2022.11166] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/09/2021] [Indexed: 12/05/2022] Open
Abstract
The aim of the present study was to investigate the expression levels of microRNA(miR)-4722-5p and miR-615-3p in Alzheimer's disease (AD) and their diagnostic value. Blood samples were collected from 33 patients with AD and 33 healthy controls, and an β-amyloid (Aβ)25-35-induced PC12 cell model was also established. The relative mRNA expression levels of miR-4722-5p and miR-615-3p were detected using reverse transcription-quantitative PCR. The correlations between the mRNA expression levels of the two miRNAs and the mini-mental state examination (MMSE) scores were analyzed, and the receiver operating characteristic curve was used to assess the diagnostic value of miR-4722-5p and miR-615-3p in AD. Functional enrichment analysis of the miRNA target genes was performed using The Database for Annotation, Visualization and Integrated Discovery database and the R language analysis package. The mRNA expression levels of miR-4722-5p and miR-615-3p were increased in patients with AD and the Aβ25-35-induced PC12 cell model. The mRNA expression levels of miR-4722-5p and miR-615-3p were negatively correlated with MMSE scores, and the combination of the two miRNAs for AD had an improved diagnostic value than that of each miRNA alone. The results of Gene Ontology (GO) enrichment analysis showed that the target genes of miR-4722-5p were found in the cytoplasm and cytosol, and were mainly involved in protein folding and cell division. The molecular functions included protein binding and GTPase activator activity. The results of Kyoto Encyclopedia of Genes and Genomes analysis showed that miR-4722-5p was associated with the regulation of dopaminergic synapses and mTOR signaling pathways. GO enrichment analysis also revealed that the target genes of miR-615-3p were located in the nucleus and cytoplasm, were involved in the regulation of transcription and protein phosphorylation, and were associated with protein binding, metal ion binding and transcription factor activity. The target genes of miR-615-3p played important roles in the regulation of the Ras and FoxO signaling pathways. In conclusion, miR-4722-5p and miR-615-3p may be potential biomarkers in the early diagnosis of AD.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Yuhao Xu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Ming Yu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
41
|
Tan Y, Cheng H, Su C, Chen P, Yang X. PI3K/Akt Signaling Pathway Ameliorates Oxidative Stress-Induced Apoptosis upon Manganese Exposure in PC12 Cells. Biol Trace Elem Res 2022; 200:749-760. [PMID: 33772736 DOI: 10.1007/s12011-021-02687-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/21/2021] [Indexed: 12/16/2022]
Abstract
Manganese (Mn)-induced neurotoxicity has aroused public concerns for many years, but its precise mechanism is still poorly understood. Herein, we report the impacts of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway in mediating neurological effects induced by manganese sulfate (MnSO4) exposure in PC12 cells. In this study, cells were treated with MnSO4 for 24 h in the absence or presence of LY294002 (a special inhibitor of PI3K). We investigated cell viability and apoptosis signals, as well as levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), and malondialdehyde (MDA). The mRNA levels of B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and Caspase-3 were also quantified through real-time quantitative PCR (RT-qPCR); protein levels of serine/threonine protein kinase (Akt) and forkhead box O3A (Foxo3a) were determined by western blot. Increasing of MnSO4 doses led to decreased SOD, GSH-Px, and CAT activities, while the level of MDA was upregulated. Moreover, cell apoptosis was significantly increased, as the mRNA of Bcl-2 and Caspase-3 was significantly decreased, while Bax mRNA was increased. Phosphorylated Akt (p-Akt) and Foxo3a (p-Foxo3a) were upregulated in a dose-dependent manner. In addition, LY294002 pretreatment reduced the activity of SOD, GSH-Px, and CAT but elevated MDA levels. Meanwhile, LY294002 pretreatment also increased cell apoptosis given the upregulated Bax and Caspase-3 mRNAs and decreased Bcl-2 mRNA. In summary, the PI3K/Akt signaling pathway can be activated by MnSO4 exposure and mediate MnSO4-induced neurotoxicity.
Collapse
Affiliation(s)
- Yanli Tan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Hong Cheng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Cheng Su
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, China.
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, China.
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, China.
| |
Collapse
|
42
|
Tariq K, Luikart BW. Striking a balance: PIP 2 and PIP 3 signaling in neuronal health and disease. EXPLORATION OF NEUROPROTECTIVE THERAPY 2022; 1:86-100. [PMID: 35098253 PMCID: PMC8797975 DOI: 10.37349/ent.2021.00008] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Phosphoinositides are membrane phospholipids involved in a variety of cellular processes like growth, development, metabolism, and transport. This review focuses on the maintenance of cellular homeostasis of phosphatidylinositol 4,5-bisphosphate (PIP2), and phosphatidylinositol 3,4,5-trisphosphate (PIP3). The critical balance of these PIPs is crucial for regulation of neuronal form and function. The activity of PIP2 and PIP3 can be regulated through kinases, phosphatases, phospholipases and cholesterol microdomains. PIP2 and PIP3 carry out their functions either indirectly through their effectors activating integral signaling pathways, or through direct regulation of membrane channels, transporters, and cytoskeletal proteins. Any perturbations to the balance between PIP2 and PIP3 signaling result in neurodevelopmental and neurodegenerative disorders. This review will discuss the upstream modulators and downstream effectors of the PIP2 and PIP3 signaling, in the context of neuronal health and disease.
Collapse
Affiliation(s)
- Kamran Tariq
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
43
|
Identification of let-7f and miR-338 as plasma-based biomarkers for sporadic amyotrophic lateral sclerosis using meta-analysis and empirical validation. Sci Rep 2022; 12:1373. [PMID: 35082326 PMCID: PMC8791978 DOI: 10.1038/s41598-022-05067-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
AbstractAmyotrophic lateral sclerosis (ALS) is a lethal neurodegenerative disease that in most cases occurs sporadic (sALS). The disease is not curable, and its pathogenesis mechanisms are not well understood yet. Given the intricacy of underlying molecular interactions and heterogeneity of ALS, the discovery of molecules contributing to disease onset and progression will open a new avenue for advancement in early diagnosis and therapeutic intervention. Here we conducted a meta-analysis of 12 circulating miRNA profiling studies using the robust rank aggregation (RRA) method, followed by enrichment analysis and experimental verification. We identified miR-451a and let-7f-5p as meta-signature miRNAs whose targets are involved in critical pathogenic pathways underlying ALS, including ‘FoxO signaling pathway’, ‘MAPK signaling pathway’, and ‘apoptosis’. A systematic review of 7 circulating gene profiling studies elucidated that 241 genes up-regulated in sALS circulation with concomitant being targets of the meta-signature miRNAs. Protein–protein interaction (PPI) network analysis of the candidate targets using MCODE algorithm revealed the main subcluster is involved in multiple cascades eventually leads apoptosis, including ‘positive regulation of neuron apoptosis. Besides, we validated the meta-analysis results using RT-qPCR. Indeed, relative expression analysis verified let-7f-5p and miR-338-3p as significantly down-regulated and up-regulated biomarkers in the plasma of sALS patients, respectively. Receiver operating characteristic (ROC) analysis also highlighted the let-7f-5p and miR-338-3p potential as robustness plasma biomarkers for diagnosis and potential therapeutic targets of sALS disease.
Collapse
|
44
|
Ji JS, Liu L, Zeng Y, Yan LL. Effect of FOXO3 and Air Pollution on Cognitive Function: A Longitudinal Cohort Study of Older Adults in China From 2000 to 2014. J Gerontol A Biol Sci Med Sci 2022; 77:1534-1541. [PMID: 35029671 PMCID: PMC9890624 DOI: 10.1093/gerona/glac016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Forkhead Box O 3 (FOXO3) genotype is strongly associated with human longevity and may be protective against neurodegeneration. Air pollution is a risk factor for cognitive decline and dementia. We aimed to study the individual and combined effects of FOXO3 and air pollution on cognitive function in a large prospective cohort with up to 14 years of follow-up. We measured cognitive function and impairment using the Mini-Mental State Examination (MMSE). We used tagging SNPs rs2253310, rs2802292, and rs4946936 to identify the FOXO3 gene, of which roughly half of the population had the longevity-associated polymorphism. We matched annual average fine particulate matter (PM2.5) concentrations within a 1 km2 grid. We conducted cross-sectional and longitudinal analyses using multivariable linear and logistic regression models and generalized estimating equations. At baseline, carriers of the longevity-associated homozygous minor alleles of FOXO3 SNPs had a higher MMSE score than the carriers of homozygous major alleles. In the longitudinal follow-up, carriers of FOXO3 homozygous minor alleles had lower odds of cognitive impairment compared with noncarriers. Higher PM2.5 was associated with a lower MMSE score and higher odds of cognitive impairment. The positive effects of FOXO3 were the strongest in females, older people, and residents in areas with lower air pollution.
Collapse
Affiliation(s)
- John S Ji
- Address correspondence to: John S. Ji, ScD, Vanke School of Public Health, Tsinghua University, 4th Floor, Mingli Building, Haidian District, Beijing 100083, China. E-mail:
| | - Linxin Liu
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Yi Zeng
- Center for Healthy Aging and Development Studies, National School of Development, Peking University, Beijing, China
- Center for the Study of Aging and Human Development, Duke Medical School, Durham, North Carolina, USA
| | - Lijing L Yan
- Global Health Research Center, Duke Kunshan University, Kunshan, China
| |
Collapse
|
45
|
Wang YQ, Jiang YJ, Zou MS, Liu J, Zhao HQ, Wang YH. Antidepressant actions of melatonin and melatonin receptor agonist: Focus on pathophysiology and treatment. Behav Brain Res 2021; 420:113724. [PMID: 34929236 DOI: 10.1016/j.bbr.2021.113724] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
Depression has become one of the most commonly prevalent neuropsychiatric disorders, and the main characteristics of depression are sleep disorders and melatonin secretion disorders caused by circadian rhythm disorders. Abnormal endogenous melatonin alterations can contribute to the occurrence and development of depression. However, molecular mechanisms underlying this abnormality remain ambiguous. The present review summarizes the mechanisms underlying the antidepressant effects of melatonin, which is related to its functions in the regulation of the hypothalamic-pituitary-adrenal axis, inhibition of neuroinflammation, inhibition of oxidative stress, alleviation of autophagy, and upregulation of neurotrophic, promotion of neuroplasticity and upregulation of the levels of neurotransmitters, etc. Also, melatonin receptor agonists, such as agomelatine, ramelteon, piromelatine, tasimelteon, and GW117, have received considerable critical attention and are highly implicated in treating depression and comorbid disorders. This review focuses on melatonin and various melatonin receptor agonists in the pathophysiology and treatment of depression, aiming to provide further insight into the pathogenesis of depression and explore potential targets for novel agent development.
Collapse
Affiliation(s)
- Ye-Qing Wang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Ya-Jie Jiang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Man-Shu Zou
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jian Liu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hong-Qing Zhao
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Yu-Hong Wang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
46
|
Lee SH, Jung S, Lee YJ, Hyun M, Chung KC. FBXO7 triggers caspase 8-mediated proteolysis of the transcription factor FOXO4 and exacerbates neuronal cytotoxicity. J Biol Chem 2021; 297:101426. [PMID: 34800438 PMCID: PMC8665361 DOI: 10.1016/j.jbc.2021.101426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 01/26/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of midbrain dopamine neurons in the substantia nigra. Mutations in the F-box only protein 7 gene (Fbxo7) have been reported to cause an autosomal recessive form of early-onset familial PD. FBXO7 is a part of the SKP1-Cullin1-F-box (SCF) E3 ubiquitin ligase complex, which mediates ubiquitination of numerous substrates. FBXO7 also regulates mitophagy, cell growth, and proteasome activity. A member of the FOXO family, the transcription factor FOXO4, is also known to modulate several cellular responses, including cell cycle progression and apoptosis; however, the relationship between FBXO7 and FOXO4 has not been investigated. In this study, we determined that FBXO7 binds to FOXO4 and negatively regulates intracellular FOXO4 levels. Interestingly, we also found that FBXO7-mediated degradation of FOXO4 did not occur through either of two major proteolysis systems, the ubiquitin-proteasome system or the lysosome-autophagy pathway, although it was blocked by a caspase 8-specific inhibitor and caspase 8-knockdown. Moreover, intracellular FOXO4 levels were greatly reduced in dopaminergic MN9D cells following treatment with neurotoxic 6-hydroxydopamine (6-OHDA), which was produced upon FBXO7-mediated and caspase 8-mediated proteolysis. Taken together, these results suggest that FOXO4 is negatively regulated in FBXO7-linked PD through caspase 8 activation, suppressing the cytoprotective effect of FOXO4 during 6-OHDA-induced neuronal cell death.
Collapse
Affiliation(s)
- Su Hyoun Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Sungyeon Jung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yun Ju Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Minju Hyun
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| |
Collapse
|
47
|
Takamatsu K, Tanaka J, Katada R, Azuma K, Takakura I, Aota K, Kamatani T, Shirota T, Inoue S, Mishima K. Aging-associated stem/progenitor cell dysfunction in the salivary glands of mice. Exp Cell Res 2021; 409:112889. [PMID: 34678306 DOI: 10.1016/j.yexcr.2021.112889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 11/25/2022]
Abstract
Although stem cell aging leads to a decline in tissue homeostasis and regenerative capacity, it remains unclear whether salivary gland stem cell function changes during this process. However, the salivary glands are gradually replaced by connective tissue during aging. Here, we show a decline in the stem cell ability of CD133-positive stem/progenitor cells in the salivary glands of aged mice. The CD133-positive cells were isolated from young, adult, and aged mice. The number of CD133-positive cells was significantly decreased in aged mice. They also showed a lower sphere formation capacity compared to young and adult mice. RNA sequencing revealed that CD133-positive cells in aged mice exhibited lower gene expression of several aging-related genes, including FoxO3a, than those in young and adult mice. Salivary gland cells infected with a recombinant lentivirus encoding the FoxO3a gene showed a reduction in oxidative stress induced by hydrogen peroxide compared with those infected with a control virus. Thus, FoxO3a may inhibit stem cell aging via oxidative stress.
Collapse
Affiliation(s)
- Koki Takamatsu
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan
| | - Junichi Tanaka
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan
| | - Ryogo Katada
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Kotaro Azuma
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Ikuko Takakura
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan; Department of Dentistry, Jikei University School of Medicine, Tokyo, Japan
| | - Keiko Aota
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takaaki Kamatani
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, Tokyo, Japan
| | - Satoshi Inoue
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan; Division of Gene Regulation and Signal Transduction, Research Center of Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan.
| |
Collapse
|
48
|
miR-223 Enhances the Neuroprotection of Estradiol Against Oxidative Stress Injury by Inhibiting the FOXO3/TXNIP Axis. Neurochem Res 2021; 47:1865-1877. [PMID: 34843004 DOI: 10.1007/s11064-021-03490-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/20/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disorder characterized by complex pathogenesis, of which oxidative stress has long been regarded as a major mechanism. Previously, the protective effects of estradiol on SH-SY5Y cells against Aβ42-induced injuries were demonstrated. In this study, the protection of SH-SY5Y cells by estradiol from H2O2-caused oxidative stress injury and Alzheimer's mice was further confirmed. H2O2 downregulated, whereas estradiol upregulated miR-223 expression. miR-223 overexpression promoted cell viability, inhibited cell apoptosis, reduced ROS levels, enhanced Superoxide Dismutase (SOD) activity, and decreased malondialdehyde (MDA) content. However, miR-223 inhibition exerted opposite effects. miR-223 directly targeted forkhead box O3 (FOXO3) and inhibited FOXO3 expression. H2O2 increased, whereas estradiol decreased thioredoxin interacting protein (TXNIP) levels; FOXO3 positively regulated TXNIP protein levels. In SH-SY5Y cells, FOXO3 overexpression increased, whereas FOXO3 knockdown reduced the cell apoptosis and ROS levels. FOXO3 bound to TXNIP promoter region and activated TXNIP transcription, whereas the activation could be partially inhibited by estradiol. Collectively, the FOXO3/TXNIP axis is downstream of miR-223. miR-223 enhances the neuroprotection of estradiol against oxidative stress injury through the FOXO3/TXNIP axis.
Collapse
|
49
|
Zhang W, Bai SS, Zhang Q, Shi RL, Wang HC, Liu YC, Ni TJ, Wu Y, Yao ZY, Sun Y, Wang MY. Physalin B reduces Aβ secretion through down-regulation of BACE1 expression by activating FoxO1 and inhibiting STAT3 phosphorylation. Chin J Nat Med 2021; 19:732-740. [PMID: 34688463 DOI: 10.1016/s1875-5364(21)60090-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Indexed: 11/18/2022]
Abstract
Physalin B (PB), one of the major active steroidal constituents of Solanaceae Physalis plants, has a wide variety of biological activities. We found that PB significantly down-regulated β-amyloid (Aβ) secretion in N2a/APPsw cells. However, the underlying mechanisms are not well understood. In the current study, we investigated the changes in key enzymes involved in β-amyloid precursor protein (APP) metabolism and other APP metabolites by treating N2a/APPsw cells with PB at different concentrations. The results indicated that PB reduced Aβ secretion, which was caused by down-regulation of β-secretase (BACE1) expression, as indicated at both the protein and mRNA levels. Further research revealed that PB regulated BACE1 expression by inducing the activation of forkhead box O1 (FoxO1) and inhibiting the phosphorylation of signal transducer and activator of transcription 3 (STAT3). In addition, the effect of PB on BACE1 expression and Aβ secretion was reversed by treatment with FoxO1 siRNA and STAT3 antagonist S3I-201. In conclusion, these data demonstrated that PB can effectively down-regulate the expression of BACE1 to reduce Aβsecretion by activating the expression of FoxO1 and inhibiting the phosphorylation of STAT3.
Collapse
Affiliation(s)
- Wei Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Shan-Shan Bai
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Central Laboratory, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang 471003, China
| | - Qi Zhang
- Grade 2019, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Ru-Ling Shi
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - He-Cheng Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - You-Cai Liu
- School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Tian-Jun Ni
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Wu
- School of Nursing Care, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhao-Yang Yao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Yi Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Ming-Yong Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China.
| |
Collapse
|
50
|
Dang R, Yang M, Cui C, Wang C, Zhang W, Geng C, Han W, Jiang P. Activation of angiotensin-converting enzyme 2/angiotensin (1-7)/mas receptor axis triggers autophagy and suppresses microglia proinflammatory polarization via forkhead box class O1 signaling. Aging Cell 2021; 20:e13480. [PMID: 34529881 PMCID: PMC8520723 DOI: 10.1111/acel.13480] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 07/03/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Brain renin‐angiotensin (Ang) system (RAS) is implicated in neuroinflammation, a major characteristic of aging process. Angiotensin (Ang) II, produced by angiotensin‐converting enzyme (ACE), activates immune system via angiotensin type 1 receptor (AT1), whereas Ang(1–7), generated by ACE2, binds with Mas receptor (MasR) to restrain excessive inflammatory response. Therefore, the present study aims to explore the relationship between RAS and neuroinflammation. We found that repeated lipopolysaccharide (LPS) treatment shifted the balance between ACE/Ang II/AT1 and ACE2/Ang(1–7)/MasR axis to the deleterious side and treatment with either MasR agonist, AVE0991 (AVE) or ACE2 activator, diminazene aceturate, exhibited strong neuroprotective actions. Mechanically, activation of ACE2/Ang(1–7)/MasR axis triggered the Forkhead box class O1 (FOXO1)‐autophagy pathway and induced superoxide dismutase (SOD) and catalase (CAT), the FOXO1‐targeted antioxidant enzymes. Meanwhile, knockdown of MasR or FOXO1 in BV2 cells, or using the selective FOXO1 inhibitor, AS1842856, in animals, suppressed FOXO1 translocation and compromised the autophagic process induced by MasR activation. We further used chloroquine (CQ) to block autophagy and showed that suppressing either FOXO1 or autophagy abrogated the anti‐inflammatory action of AVE. Likewise, Ang(1–7) also induced FOXO1 signaling and autophagic flux following LPS treatment in BV2 cells. Cotreatment with AS1842856 or CQ all led to autophagic inhibition and thereby abolished Ang(1–7)‐induced remission on NLRP3 inflammasome activation caused by LPS exposure, shifting the microglial polarization from M1 to M2 phenotype. Collectively, these results firstly illustrated the mechanism of ACE2/Ang(1–7)/MasR axis in neuroinflammation, strongly indicating the involvement of FOXO1‐mediated autophagy in the neuroimmune‐modulating effects triggered by MasR activation.
Collapse
Affiliation(s)
- Ruili Dang
- Institute of Clinical Pharmacy and Pharmacology Jining First People’s HospitalJining Medical University Jining China
| | - Mengqi Yang
- Institute of Clinical Pharmacy and Pharmacology Jining First People’s HospitalJining Medical University Jining China
| | - Changmeng Cui
- Department of Neurosurgery Affiliated Hospital of Jining Medical University Jining China
| | - Changshui Wang
- Department of Neurosurgery Affiliated Hospital of Jining Medical University Jining China
| | - Wenyuan Zhang
- Department of Pharmacy Zhongshan Affiliated Hospital of Zhongshan University Zhongshan China
| | - Chunmei Geng
- Institute of Clinical Pharmacy and Pharmacology Jining First People’s HospitalJining Medical University Jining China
| | - Wenxiu Han
- Institute of Clinical Pharmacy and Pharmacology Jining First People’s HospitalJining Medical University Jining China
| | - Pei Jiang
- Institute of Clinical Pharmacy and Pharmacology Jining First People’s HospitalJining Medical University Jining China
| |
Collapse
|