1
|
Zhou KG, Huang YB, Zhu ZW, Jiang M, Jin LJ, Guan Q, Tian LL, Zhang JX. Mesencephalic astrocyte-derived neurotrophic factor inhibits neuroinflammation through autophagy-mediated α-synuclein degradation. Arch Gerontol Geriatr 2025; 131:105738. [PMID: 39761611 DOI: 10.1016/j.archger.2024.105738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder marked by the progressive loss of dopamine neurons in the substantia nigra. α-synuclein (SNCA) aggregation-induced microglia activation and neuroinflammation play vital role in the pathology of PD. Our previous studies showed that mesencephalic astrocyte-derived neurotrophic factor (MANF) could inhibit SNCA accumulation and Lipopolysaccharides (LPS)-induced neuroinflammation, but the specific molecular mechanism remains unclear. In this study, we showed that knock-down the expression of MANF leads to the up-regulation of inflammatory factor tumor necrosis factor-α (TNF-α). Exogenous MANF protein inhibits LPS-induced neuroinflammation in BV2 cells. Additionally, our results indicated that knock-down of the expression of MANF triggered autophagic pathway dysfunction, while exogenous addition of MANF protein or using adeno-associated virus 8 (AAV8) mediated MANF over-expression could activate the autophagic system and subsequently suppress SNCA accumulation. Furthermore, using autophagy inhibitor to block autophagic flux, we found that MANF prevented neuroinflammation by autophagy-mediated SNCA degradation. Collectively, this study indicated that MANF has potential therapeutic value for PD. Autophagy and its role in MANF-mediated anti-inflammatory properties may provide new sights that target SNCA pathology in PD.
Collapse
Affiliation(s)
- Kai-Ge Zhou
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yi-Bin Huang
- School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zi-Wen Zhu
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Ming Jiang
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Biomedical Research Center, Tongji University Suzhou Institute, Jiangsu, 215101, China
| | - Ling-Jing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qiang Guan
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lu-Lu Tian
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Jing-Xing Zhang
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
2
|
Qiu D, Wang L, Wang L, Dong Y. Human platelet lysate: a potential therapeutic for intracerebral hemorrhage. Front Neurosci 2025; 18:1517601. [PMID: 39881806 PMCID: PMC11774881 DOI: 10.3389/fnins.2024.1517601] [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: 10/26/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is a major public health challenge worldwide, and is associated with elevated rates of mortality, disability, and morbidity, especially in low- and middle-income nations. However, our knowledge of the detailed molecular processes involved in ICH remains insufficient, particularly those involved in the secondary injury stage, resulting in a lack of effective treatments for ICH. Human platelet lysates (HPL) are abundant in bioactive factors, and numerous studies have demonstrated their beneficial effects on neurological diseases, including their anti-neuroinflammatory ability, anti-oxidant effects, maintenance of blood-brain barrier integrity, and promotion of neurogenesis. In this review, we thoroughly explore the potential of HPL for treating ICH from three critical perspectives: the rationale for selecting HPL as a treatment for ICH, the mechanisms through which HPL contributes to ICH management, and the additional measures necessary for HPL as a treatment for ICH. We elucidate the role of platelets in ICH pathophysiology and highlight the limitations of the current treatment options and advancements in preclinical research on the application of HPL in neurological disorders. Furthermore, historical developments and preparation methods of HPL in the field of biomedicine are discussed. Additionally, we summarize the bioactive molecules present in HPL and their potential therapeutic effects in ICH. Finally, we outline the issues that must be addressed regarding utilizing HPL as a treatment modality for ICH.
Collapse
Affiliation(s)
- Dachang Qiu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lin Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lanlan Wang
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yongfei Dong
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
3
|
Di Menna L, Alborghetti M, De Bartolo MI, Borro M, Gentile G, Zinni M, Bologna M, Cutrona C, D'Errico G, Imbriglio T, Bucci D, Merlo S, Ginerete RP, Orlando R, Carrillo F, Fortunato G, Cannella M, Sortino MA, Pansiot J, Baud O, Nicoletti F, Bruno V, Simmaco M, Pontieri FE, Bianchini E, Rinaldi D, de Curtis A, De Gaetano G, Iacoviello L, Esposito T, Berardelli A, Battaglia G. Preclinical and clinical study on type 3 metabotropic glutamate receptors in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:9. [PMID: 39755677 DOI: 10.1038/s41531-024-00860-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/22/2024] [Indexed: 01/06/2025] Open
Abstract
Metabotropic glutamate (mGlu) receptors are candidate drug targets for therapeutic intervention in Parkinson's disease (PD). Here we focused on mGlu3, a receptor subtype involved in synaptic regulation and neuroinflammation. mGlu3-/- mice showed an enhanced nigro-striatal damage and microglial activation in response to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Expression of genes encoding anti-inflammatory proteins and neuroprotective factors was reduced in the striatum of MPTP-treated mGlu3-/- mice. We also examined polymorphic variants of GRM3 (the mGlu3 receptor encoding gene) in 723 PD patients and 826 healthy controls. Two GRM3 haplotypes were associated with PD, and gene variants correlated with motor and non-motor signs. Interestingly, PD patients carrying each of the two haplotypes showed an impaired cortical plasticity in the paired associated stimulation paradigm of magnetic transcranial stimulation. These findings suggest that mGlu3 receptors are neuroprotective in mouse models of parkinsonism and shape mechanisms of cortical plasticity in PD.
Collapse
Grants
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- RF-2018-123-68289 Ministero della Salute (Ministry of Health, Italy)
- E64117000190001 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- E64117000190001 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- E64117000190001 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- E64117000190001 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- E64117000190001 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
Collapse
Affiliation(s)
- Luisa Di Menna
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
| | - Marika Alborghetti
- Departments of Human Neurosciences, Sapienza University of Rome, Rome, Italy
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
| | | | - Marina Borro
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
| | - Giovanna Gentile
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
| | - Manuela Zinni
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Matteo Bologna
- Departments of Human Neurosciences, Sapienza University of Rome, Rome, Italy
- Department of Clinical Neurophysiology, IRCCS Neuromed, Pozzilli, Italy
| | - Carolina Cutrona
- Departments of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | | | | | - Domenico Bucci
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
| | - Sara Merlo
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | | | - Federica Carrillo
- Institute of Genetics and Biophysics, Adriano Buzzati-Traverso, National Research Council, Naples, Italy
| | - Giorgio Fortunato
- Institute of Genetics and Biophysics, Adriano Buzzati-Traverso, National Research Council, Naples, Italy
| | - Milena Cannella
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Julien Pansiot
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
| | - Olivier Baud
- Inserm UMR1141 NeuroDiderot, Université Paris Cité, Paris, France
- Department of Neonatal Medicine, Cochin-Port Royal Hospital, FHU PREMA, AP-HP Centre, Paris, France
- Université Paris Cite, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPE, French Institute for Medical Research and Health INSERM, INRAE, 75014, Paris, France
| | - Ferdinando Nicoletti
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Valeria Bruno
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Maurizio Simmaco
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
| | - Francesco Ernesto Pontieri
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
| | - Edoardo Bianchini
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
- AGEIS, Université Grenoble Alpes, 38000, Grenoble, France
| | - Domiziana Rinaldi
- Neuroscience, Mental Health, and Sensory Organs Clinical Neurophysiology, Sapienza University of Rome, Rome, Italy
| | - Amalia de Curtis
- Department of Epidemiology and Prevention, IRCCS Neuromed, 86077, Pozzilli, Italy
| | - Giovanni De Gaetano
- Department of Epidemiology and Prevention, IRCCS Neuromed, 86077, Pozzilli, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS Neuromed, 86077, Pozzilli, Italy
- Department of Medicine and Surgery, LUM University, 70010, Casamassima, Bari, Italy
| | - Teresa Esposito
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
- Institute of Genetics and Biophysics, Adriano Buzzati-Traverso, National Research Council, Naples, Italy
| | - Alfredo Berardelli
- Departments of Human Neurosciences, Sapienza University of Rome, Rome, Italy.
- Department of Clinical Neurophysiology, IRCCS Neuromed, Pozzilli, Italy.
| | - Giuseppe Battaglia
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
4
|
Kim J, Chang MY. Gene Therapy for Parkinson's Disease Using Midbrain Developmental Genes to Regulate Dopaminergic Neuronal Maintenance. Int J Mol Sci 2024; 25:12369. [PMID: 39596436 PMCID: PMC11594980 DOI: 10.3390/ijms252212369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. It is characterized by the progressive loss of dopaminergic (DAnergic) neurons in the substantia nigra and decreased dopamine (DA) levels, which lead to both motor and non-motor symptoms. Conventional PD treatments aim to alleviate symptoms, but do not delay disease progression. PD gene therapy offers a promising approach to improving current treatments, with the potential to alleviate significant PD symptoms and cause fewer adverse effects than conventional therapies. DA replacement approaches and DA enzyme expression do not slow disease progression. However, DA replacement gene therapies, such as adeno-associated virus (AAV)-glutamic acid decarboxylase (GAD) and L-amino acid decarboxylase (AADC) gene therapies, which increase DA transmitter levels, have been demonstrated to be safe and efficient in early-phase clinical trials. Disease-modifying strategies, which aim to slow disease progression, appear to be potent. These include therapies targeting downstream pathways, neurotrophic factors, and midbrain DAnergic neuronal factors, all of which have shown potential in preclinical and clinical trials. These approaches focus on maintaining the integrity of DAnergic neurons, not just targeting the DA transmitter level itself. In particular, critical midbrain developmental and maintenance factors, such as Nurr1 and Foxa2, can interact synergistically with neighboring glia, in a paracrine mode of action, to protect DAnergic neurons against various toxic factors. Similar outcomes could be achieved by targeting both DAnergic neurons and glial cells with other candidate gene therapies, but in-depth research is needed. Neurotrophic factors, such as neurturin, the glial-cell-line-derived neurotrophic factor (GDNF), the brain-derived neurotrophic factor (BDNF), and the vascular endothelial growth factor (VEGF), are also being investigated for their potential to support DAnergic neuron survival. Additionally, gene therapies targeting key downstream pathways, such as the autophagy-lysosome pathway, mitochondrial function, and endoplasmic reticulum (ER) stress, offer promising avenues. Gene editing and delivery techniques continue to evolve, presenting new opportunities to develop effective gene therapies for PD.
Collapse
Affiliation(s)
- Jintae Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Republic of Korea;
| | - Mi-Yoon Chang
- Department of Premedicine, College of Medicine, Hanyang University, FTC12, 222 Wangsimni-ro, Seoul 04763, Republic of Korea
- Biomedical Research Institute, Hanyang University, Seoul 04763, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
5
|
Shen Y, Wang J, Liang J, Chen Y, Wu X, Ren Z, Zhou J, Feng L, Shen Y. E3 Ubiquitin Ligase Ring Finger Protein 2 Alleviates Cerebral Ischemia-Reperfusion Injury by Stabilizing Mesencephalic Astrocyte-Derived Neurotrophic Factor Through Monoubiquitination. CNS Neurosci Ther 2024; 30:e70136. [PMID: 39614674 DOI: 10.1111/cns.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 12/01/2024] Open
Abstract
AIM Cerebral ischemic stroke (IS) is one of the leading causes of morbidity and mortality globally. However, the mechanisms underlying IS injury remain poorly understood. Ring finger protein 2 (RNF2), the member of the polycomb family (PcG), has been implicated in diverse biological and pathological conditions. However, whether RNF2 plays a role in IS progression is not clarified. This study aims to investigate the potential effects of RNF2 on IS. METHODS The effects of RNF2 were studied in human postmortem IS brains, a rat model of IS, tunicamycin (TM)-induced mouse neuroblastoma neuro2a (N2a) cells, and oxygen-glucose deprivation/reperfusion (OGD/R)-induced SH-SY5Y cells. RESULTS Here, we demonstrated that RNF2 was markedly upregulated both in human postmortem IS brains and ischemic rat brains and RNF2 overexpression alleviated brain injury induced by middle cerebral artery occlusion by reducing neuron apoptosis. Mechanistically, we found that RNF2 is an E3 ubiquitin ligase for the mesencephalic astrocyte-derived neurotrophic factor (MANF), which confers protection against brain ischemia. RNF2 interacted with MANF and promoted the monoubiquitination of MANF, consequently facilitating its stability and nuclear localization. CONCLUSION Collectively, RNF2 is identified as a critical inhibitor of IS injury by stabilizing MANF through monoubiquitination, suggesting that RNF2 is a potential therapeutic target for IS.
Collapse
Affiliation(s)
- Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
| | - Jinfeng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
| | - Junxing Liang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
| | - Ying Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
| | - Xueyan Wu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Anhui Medical University, Hefei, China
| | - Zhenhua Ren
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Anhui Medical University, Hefei, China
| | - Jiangning Zhou
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Anhui Medical University, Hefei, China
| | - Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Anhui Medical University, Hefei, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Graewert MA, Volkova M, Jonasson K, Määttä JAE, Gräwert T, Mamidi S, Kulesskaya N, Evenäs J, Johnsson RE, Svergun D, Bhattacharjee A, Huttunen HJ. Structural basis of CDNF interaction with the UPR regulator GRP78. Nat Commun 2024; 15:8175. [PMID: 39289391 PMCID: PMC11408689 DOI: 10.1038/s41467-024-52478-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotrophic factor that is a disease-modifying drug candidate for Parkinson's disease. CDNF has pleiotropic protective effects on stressed cells, but its mechanism of action remains incompletely understood. Here, we use state-of-the-art advanced structural techniques to resolve the structural basis of CDNF interaction with GRP78, the master regulator of the unfolded protein response (UPR) pathway. Subsequent binding studies confirm the obtained structural model of the complex, eventually revealing the interaction site of CDNF and GRP78. Finally, mutating the key residues of CDNF mediating its interaction with GRP78 not only results in impaired binding of CDNF but also abolishes the neuroprotective activity of CDNF-derived peptides in mesencephalic neuron cultures. These results suggest that the molecular interaction with GRP78 mediates the neuroprotective actions of CDNF and provide a structural basis for development of next generation CDNF-based therapeutic compounds against neurodegenerative diseases.
Collapse
Affiliation(s)
- Melissa A Graewert
- European Molecular Biological Laboratory, DE-22607, Hamburg, Germany
- BIOSAXS GmbH, DE-22607, Hamburg, Germany
| | - Maria Volkova
- Red Glead Discovery AB, Medicon Village SE-223 81, Lund, Sweden
| | - Klara Jonasson
- Red Glead Discovery AB, Medicon Village SE-223 81, Lund, Sweden
| | - Juha A E Määttä
- Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33520, Finland
| | | | - Samara Mamidi
- Red Glead Discovery AB, Medicon Village SE-223 81, Lund, Sweden
| | | | - Johan Evenäs
- Red Glead Discovery AB, Medicon Village SE-223 81, Lund, Sweden
| | | | | | | | | |
Collapse
|
7
|
Zhang Y, Liu L, Qi Y, Lou J, Chen Y, Liu C, Li H, Chang X, Hu Z, Li Y, Zhang Y, Feng C, Zhou Y, Zhai Y, Li C. Lactic acid promotes nucleus pulposus cell senescence and corresponding intervertebral disc degeneration via interacting with Akt. Cell Mol Life Sci 2024; 81:24. [PMID: 38212432 PMCID: PMC11071984 DOI: 10.1007/s00018-023-05094-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024]
Abstract
The accumulation of metabolites in the intervertebral disc is considered an important cause of intervertebral disc degeneration (IVDD). Lactic acid, which is a metabolite that is produced by cellular anaerobic glycolysis, has been proven to be closely associated with IVDD. However, little is known about the role of lactic acid in nucleus pulposus cells (NPCs) senescence and oxidative stress. The aim of this study was to investigate the effect of lactic acid on NPCs senescence and oxidative stress as well as the underlying mechanism. A puncture-induced disc degeneration (PIDD) model was established in rats. Metabolomics analysis revealed that lactic acid levels were significantly increased in degenerated intervertebral discs. Elimination of excessive lactic acid using a lactate oxidase (LOx)-overexpressing lentivirus alleviated the progression of IVDD. In vitro experiments showed that high concentrations of lactic acid could induce senescence and oxidative stress in NPCs. High-throughput RNA sequencing results and bioinformatic analysis demonstrated that the induction of NPCs senescence and oxidative stress by lactic acid may be related to the PI3K/Akt signaling pathway. Further study verified that high concentrations of lactic acid could induce NPCs senescence and oxidative stress by interacting with Akt and regulating its downstream Akt/p21/p27/cyclin D1 and Akt/Nrf2/HO-1 pathways. Utilizing molecular docking, site-directed mutation and microscale thermophoresis assays, we found that lactic acid could regulate Akt kinase activity by binding to the Lys39 and Leu52 residues in the PH domain of Akt. These results highlight the involvement of lactic acid in NPCs senescence and oxidative stress, and lactic acid may become a novel potential therapeutic target for the treatment of IVDD.
Collapse
Affiliation(s)
- Yuyao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Libangxi Liu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
- Department of Orthopedics, General Hospital of Central Theater Command of PLA, Wuhan, 430000, China
| | - Yuhan Qi
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, 100000, China
| | - Jinhui Lou
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yuxuan Chen
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Chao Liu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Haiyin Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Xian Chang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Zhilei Hu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yueyang Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yang Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Chencheng Feng
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China
| | - Yu Zhai
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China.
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
8
|
Bhushan B, Singh NK. Role of Astrogliosis in the Pathogenesis of Parkinson's Disease: Insights into Astrocytic Nrf2 Pathway as a Potential Therapeutic Target. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1015-1029. [PMID: 37817521 DOI: 10.2174/0118715273270473231002104610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/21/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023]
Abstract
Recently, Parkinson's disease (PD) has become a remarkable burden on families and society with an acceleration of population aging having several pathological hallmarks such as dopaminergic neuronal loss of the substantia nigra pars compacta, α-synucleinopathy, neuroinflammation, autophagy, last but not the least astrogliosis. Astrocyte, star-shaped glial cells perform notable physiological functions in the brain through several molecular and cellular mechanisms including nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. It has been well established that the downregulation of the astrocytic Nrf2 signaling pathway plays a crucial role in the pathogenesis of PD because it is a master regulator of cellular defense mechanism along with a regulator of numerous detoxifying and antioxidant enzymes gene expression. Fascinatingly, upregulation of the astrocytic Nrf2 signaling pathway attenuates the degeneration of nigrostriatal neurons, restores neuronal proliferation, rejuvenates astrocytic functions, and exhibits neuroprotective effects via numerous cellular and molecular mechanisms in the PD-like brain of the experimental animal. Here, we discuss the numerous in-vitro and in-vivo studies that evaluate the neuroprotective potential of the astrocytic Nrf2 signaling pathway against experimentally-induced PD-like manifestation. In conclusion, based on available preclinical reports, it can be assumed that the astrocytic Nrf2 signaling pathway could be an alternative target in the drug discovery process for the prevention, management, and treatment of PD.
Collapse
Affiliation(s)
- Bharat Bhushan
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, NH-19, Mathura-Delhi Road, Chaumuhan, Mathura 281406, U.P. India
| | - Niraj Kumar Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, NH-19, Mathura-Delhi Road, Chaumuhan, Mathura 281406, U.P. India
| |
Collapse
|
9
|
Xiao Y, Zhao L, Zang X, Xue S. Compressed primary-to-transmodal gradient is accompanied with subcortical alterations and linked to neurotransmitters and cellular signatures in major depressive disorder. Hum Brain Mapp 2023; 44:5919-5935. [PMID: 37688552 PMCID: PMC10619397 DOI: 10.1002/hbm.26485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/18/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Major depressive disorder (MDD) has been shown to involve widespread changes in low-level sensorimotor and higher-level cognitive functions. Recent research found that a primary-to-transmodal gradient could capture a cortical hierarchical organization ranging from perception and action to cognition in healthy subjects, but a prominent gradient dysfunction in MDD patients. However, whether and how this cortical gradient is linked to subcortical impairments and whether it is reflected in the microscale neurotransmitter systems and cell type-specific transcriptional signatures remain largely unknown. Data were acquired from 323 MDD patients and 328 sex- and age-matched healthy controls derived from the REST-meta-MDD project, and the human brain neurotransmitter systems density maps and gene expression data were drawn from two publicly available datasets. We investigated alterations of the primary-to-transmodal gradient in MDD patients and their correlations with clinical symptoms of depression and anxiety, as well as their paralleled subcortical impairments. The correlations between MDD-related gradient alterations and densities of the neurotransmitter systems and gene expression information were assessed, respectively. The results demonstrated that MDD patients had a compressed primary-to-transmodal gradient accompanied by paralleled alterations in subcortical regions including the caudate, amygdala, and thalamus. The case-control gradient differences were spatially correlated with the densities of the neurotransmitter systems including the serotonin and dopamine receptors, and meanwhile with gene expression enriched in astrocytes, excitatory and inhibitory neuronal cells. These findings mapped the paralleled subcortical impairments in cortical hierarchical organization and also helped us understand the possible molecular and cellular substrates of the co-occurrence of high-level cognitive impairments with low-level sensorimotor abnormalities in MDD.
Collapse
Affiliation(s)
- Yang Xiao
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| | - Lei Zhao
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| | - Xuelian Zang
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| | - Shao‐Wei Xue
- Center for Cognition and Brain DisordersThe Affiliated Hospital of Hangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Institute of Psychological ScienceHangzhou Normal UniversityHangzhouZhejiang ProvincePR China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive ImpairmentsHangzhouZhejiang ProvincePR China
| |
Collapse
|
10
|
Zhang C, Zhang M, Cao X, Jiao B, Zhang W, Yu S, Zhang X. Navigating the Landscape of MANF Research: A Scientometric Journey with CiteSpace Analysis. Cell Mol Neurobiol 2023; 43:3897-3913. [PMID: 37751132 PMCID: PMC10661837 DOI: 10.1007/s10571-023-01412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/09/2023] [Indexed: 09/27/2023]
Abstract
This study employs bibliometric analysis through CiteSpace to comprehensively evaluate the status and trends of MANF (mesencephalic astrocyte-derived neurotrophic factor) research spanning 25 years (1997-2022). It aims to fill the gap in objective and comprehensive reviews of MANF research. MANF-related studies were extracted from the Web of Science database. MANF publications were quantitatively and qualitatively analyzed for various factors by CiteSpace, including publication volume, journals, countries/regions, institutions, and authors. Keywords and references were visually analyzed to unveil research evolution and hotspot. Analysis of 353 MANF-related articles revealed escalating annual publications, indicating growing recognition of MANF's importance. High-impact journals such as the International Journal of Molecular Sciences and Journal of Biological Chemistry underscored MANF's interdisciplinary significance. Collaborative networks highlighted China and the USA's pivotal roles, while influential figures and partnerships drove understanding of MANF's mechanisms. Co-word analysis of MANF-related keywords exposed key evolutionary hotspots, encompassing neurotrophic effects, cytoprotective roles, MANF-related diseases, and the CDNF/MANF family. This progression from basic understanding to clinical potential showcased MANF's versatility from cellular protection to therapy. Bibliometric analysis reveals MANF's diverse research trends and pathways, from basics to clinical applications, driving medical progress. This comprehensive assessment enriches understanding and empowers researchers for dynamic evolution, advancing innovation, and benefiting patients. Bibliometric analysis of MANF research. The graphical abstract depicts the bibliometric analysis of MANF research, highlighting its aims, methods, and key results.
Collapse
Affiliation(s)
- Caixia Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Mi Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Xueqin Cao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Bo Jiao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Wencui Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Shangchen Yu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
11
|
Wang T, Sun Y, Dettmer U. Astrocytes in Parkinson's Disease: From Role to Possible Intervention. Cells 2023; 12:2336. [PMID: 37830550 PMCID: PMC10572093 DOI: 10.3390/cells12192336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons. While neuronal dysfunction is central to PD, astrocytes also play important roles, both positive and negative, and such roles have not yet been fully explored. This literature review serves to highlight these roles and how the properties of astrocytes can be used to increase neuron survivability. Astrocytes normally have protective functions, such as releasing neurotrophic factors, metabolizing glutamate, transferring healthy mitochondria to neurons, or maintaining the blood-brain barrier. However, in PD, astrocytes can become dysfunctional and contribute to neurotoxicity, e.g., via impaired glutamate metabolism or the release of inflammatory cytokines. Therefore, astrocytes represent a double-edged sword. Restoring healthy astrocyte function and increasing the beneficial effects of astrocytes represents a promising therapeutic approach. Strategies such as promoting neurotrophin release, preventing harmful astrocyte reactivity, or utilizing regional astrocyte diversity may help restore neuroprotection.
Collapse
Affiliation(s)
- Tianyou Wang
- Collège Jean-de-Brébeuf, 3200 Chemin de la Côte-Sainte-Catherine, Montreal, QC H3T 1C1, Canada
| | - Yingqi Sun
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK;
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
12
|
Wen W, Wang Y, Li H, Hu D, Zhang Z, Lin H, Luo J. Upregulation of mesencephalic astrocyte-derived neurotrophic factor (MANF) expression offers protection against alcohol neurotoxicity. J Neurochem 2023; 166:943-959. [PMID: 37507360 PMCID: PMC10906989 DOI: 10.1111/jnc.15921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023]
Abstract
Alcohol exposure has detrimental effects on both the developing and mature brain. Endoplasmic reticulum (ER) stress is one of the mechanisms that contributes to alcohol-induced neuronal damages. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an ER stress-responsive protein and is neuroprotective in multiple neuronal injury and neurodegenerative disease models. MANF deficiency has been shown to exacerbate alcohol-induced ER stress and neurodegeneration. However, it is unknown whether MANF supplement is sufficient to protect against alcohol neurotoxicity. Alcohol alters MANF expression in the brain, but the mechanisms underlying alcohol modulation of MANF expression remain unclear. This study was designed to determine how alcohol alters MANF expression in neuronal cells and whether exogeneous MANF can alleviate alcohol neurotoxicity. We showed that alcohol increased MANF transcription and secretion without affecting MANF mRNA stability and protein degradation. ER stress was necessary for alcohol-induced MANF upregulation, as pharmacological inhibition of ER stress by 4-PBA diminished alcohol-induced MANF expression. In addition, the presence of ER stress response element II (ERSE-II) was required for alcohol-stimulated MANF transcription. Mutations or deletion of this sequence abolished alcohol-regulated transcriptional activity. We generated MANF knockout (KO) neuronal cells using CRISPR/Cas9. MANF KO cells exhibited increased unfolded protein response (UPR) and were more susceptible to alcohol-induced cell death. On the other hand, MANF upregulation by the addition of recombinant MANF protein or adenovirus gene transduction protected neuronal cells against alcohol-induced cell death. Further studies using early postnatal mouse pups demonstrated that enhanced MANF expression in the brain by intracerebroventricular (ICV) injection of MANF adeno-associated viruses ameliorated alcohol-induced cell death. Thus, alcohol increased MANF expression through inducing ER stress, which could be a protective response. Exogenous MANF was able to protect against alcohol-induced neurodegeneration.
Collapse
Affiliation(s)
- Wen Wen
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Yongchao Wang
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37372, USA
| | - Hui Li
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Di Hu
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zuohui Zhang
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Hong Lin
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Jia Luo
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- VA Iowa City Health Care System, Iowa City, IA 52246, USA
| |
Collapse
|
13
|
Siwecka N, Saramowicz K, Galita G, Rozpędek-Kamińska W, Majsterek I. Inhibition of Protein Aggregation and Endoplasmic Reticulum Stress as a Targeted Therapy for α-Synucleinopathy. Pharmaceutics 2023; 15:2051. [PMID: 37631265 PMCID: PMC10459316 DOI: 10.3390/pharmaceutics15082051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
α-synuclein (α-syn) is an intrinsically disordered protein abundant in the central nervous system. Physiologically, the protein regulates vesicle trafficking and neurotransmitter release in the presynaptic terminals. Pathologies related to misfolding and aggregation of α-syn are referred to as α-synucleinopathies, and they constitute a frequent cause of neurodegeneration. The most common α-synucleinopathy, Parkinson's disease (PD), is caused by abnormal accumulation of α-syn in the dopaminergic neurons of the midbrain. This results in protein overload, activation of endoplasmic reticulum (ER) stress, and, ultimately, neural cell apoptosis and neurodegeneration. To date, the available treatment options for PD are only symptomatic and rely on dopamine replacement therapy or palliative surgery. As the prevalence of PD has skyrocketed in recent years, there is a pending issue for development of new disease-modifying strategies. These include anti-aggregative agents that target α-syn directly (gene therapy, small molecules and immunization), indirectly (modulators of ER stress, oxidative stress and clearance pathways) or combine both actions (natural compounds). Herein, we provide an overview on the characteristic features of the structure and pathogenic mechanisms of α-syn that could be targeted with novel molecular-based therapies.
Collapse
Affiliation(s)
| | | | | | | | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (N.S.); (K.S.); (G.G.); (W.R.-K.)
| |
Collapse
|
14
|
Pakarinen E, Lindholm P. CDNF and MANF in the brain dopamine system and their potential as treatment for Parkinson's disease. Front Psychiatry 2023; 14:1188697. [PMID: 37555005 PMCID: PMC10405524 DOI: 10.3389/fpsyt.2023.1188697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/23/2023] [Indexed: 08/10/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by gradual loss of midbrain dopamine neurons, leading to impaired motor function. Preclinical studies have indicated cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF) to be potential therapeutic molecules for the treatment of PD. CDNF was proven to be safe and well tolerated when tested in Phase I-II clinical trials in PD patients. Neuroprotective and neurorestorative effects of CDNF and MANF were demonstrated in animal models of PD, where they promoted the survival of dopamine neurons and improved motor function. However, biological roles of endogenous CDNF and MANF proteins in the midbrain dopamine system have been less clear. In addition to extracellular trophic activities, CDNF/MANF proteins function intracellularly in the endoplasmic reticulum (ER), where they modulate protein homeostasis and protect cells against ER stress by regulating the unfolded protein response (UPR). Here, our aim is to give an overview of the biology of endogenous CDNF and MANF in the brain dopamine system. We will discuss recent studies on CDNF and MANF knockout animal models, and effects of CDNF and MANF in preclinical models of PD. To elucidate possible roles of CDNF and MANF in human biology, we will review CDNF and MANF tissue expression patterns and regulation of CDNF/MANF levels in human diseases. Finally, we will discuss novel findings related to the molecular mechanism of CDNF and MANF action in ER stress, UPR, and inflammation, all of which are mechanisms potentially involved in the pathophysiology of PD.
Collapse
Affiliation(s)
| | - Päivi Lindholm
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Sousa NS, Brás MF, Antunes IB, Lindholm P, Neves J, Sousa-Victor P. Aging disrupts MANF-mediated immune modulation during skeletal muscle regeneration. NATURE AGING 2023; 3:585-599. [PMID: 37118549 DOI: 10.1038/s43587-023-00382-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/16/2023] [Indexed: 04/30/2023]
Abstract
Age-related decline in skeletal muscle regenerative capacity is multifactorial, yet the contribution of immune dysfunction to regenerative failure is unknown. Macrophages are essential for effective debris clearance and muscle stem cell activity during muscle regeneration, but the regulatory mechanisms governing macrophage function during muscle repair are largely unexplored. Here, we uncover a new mechanism of immune modulation operating during skeletal muscle regeneration that is disrupted in aged animals and relies on the regulation of macrophage function. The immune modulator mesencephalic astrocyte-derived neurotrophic factor (MANF) is induced following muscle injury in young mice but not in aged animals, and its expression is essential for regenerative success. Regenerative impairments in aged muscle are associated with defects in the repair-associated myeloid response similar to those found in MANF-deficient models and could be improved through MANF delivery. We propose that restoring MANF levels is a viable strategy to improve myeloid response and regenerative capacity in aged muscle.
Collapse
Affiliation(s)
- Neuza S Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Margarida F Brás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Inês B Antunes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Päivi Lindholm
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Joana Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Sousa-Victor
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
16
|
Kim S, Pajarillo E, Nyarko-Danquah I, Aschner M, Lee E. Role of Astrocytes in Parkinson's Disease Associated with Genetic Mutations and Neurotoxicants. Cells 2023; 12:622. [PMID: 36831289 PMCID: PMC9953822 DOI: 10.3390/cells12040622] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons and the aggregation of Lewy bodies in the basal ganglia, resulting in movement impairment referred to as parkinsonism. However, the etiology of PD is not well known, with genetic factors accounting only for 10-15% of all PD cases. The pathogenetic mechanism of PD is not completely understood, although several mechanisms, such as oxidative stress and inflammation, have been suggested. Understanding the mechanisms of PD pathogenesis is critical for developing highly efficacious therapeutics. In the PD brain, dopaminergic neurons degenerate mainly in the basal ganglia, but recently emerging evidence has shown that astrocytes also significantly contribute to dopaminergic neuronal death. In this review, we discuss the role of astrocytes in PD pathogenesis due to mutations in α-synuclein (PARK1), DJ-1 (PARK7), parkin (PARK2), leucine-rich repeat kinase 2 (LRRK2, PARK8), and PTEN-induced kinase 1 (PINK1, PARK6). We also discuss PD experimental models using neurotoxins, such as paraquat, rotenone, 6-hydroxydopamine, and MPTP/MPP+. A more precise and comprehensive understanding of astrocytes' modulatory roles in dopaminergic neurodegeneration in PD will help develop novel strategies for effective PD therapeutics.
Collapse
Affiliation(s)
- Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|