1
|
Shi Z, Mao L, Chen S, Du Z, Xiang J, Shi M, Wang Y, Wang Y, Chen X, Xu Z, Gao Y. Reversing Persistent PTEN Activation after Traumatic Brain Injury Fuels Long-Term Axonal Regeneration via Akt/mTORC1 Signaling Cascade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410136. [PMID: 39680734 PMCID: PMC11809353 DOI: 10.1002/advs.202410136] [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: 08/23/2024] [Revised: 11/24/2024] [Indexed: 12/18/2024]
Abstract
Traumatic brain injury (TBI) often leads to enduring axonal damage and persistent neurological deficits. While PTEN's role in neuronal growth is recognized, its long-term activation changes post-TBI and its effects on sensory-motor circuits are not well understood. Here, it is demonstrated that the neuronal knockout of PTEN (PTEN-nKO) significantly enhances both structural and functional recovery over the long term after TBI. Importantly, in vivo, DTI-MRI revealed that PTEN-nKO promotes white matter repair post-TBI. Additionally, calcium imaging and electromyographic recordings indicated that PTEN-nKO facilitates cortical remapping and restores sensory-motor pathways. Mechanistically, PTEN negatively regulates the Akt/mTOR pathway by inhibiting Akt, thereby suppressing mTOR. Raptor is a key component of mTORC1 and its suppression impedes axonal regeneration. The restoration of white matter integrity and the improvements in neural function observed in PTEN-nKO TBI-treated mice are reversed by a PTEN/Raptor double knockout (PTEN/Raptor D-nKO), suggesting that mTORC1 acts as a key mediator. These findings highlight persistent alterations in the PTEN/Akt/mTORC1 axis are critical for neural circuit remodeling and cortical remapping post-TBI, offering new insights into TBI pathophysiology and potential therapeutic targets.
Collapse
Affiliation(s)
- Ziyu Shi
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Leilei Mao
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Shuning Chen
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Zhuoying Du
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Jiakun Xiang
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Minghong Shi
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yana Wang
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yuqing Wang
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Xingdong Chen
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Zhi‐Xiang Xu
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yanqin Gao
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
2
|
Mi Z, Povysheva N, Rose ME, Ma J, Zeh DJ, Harikumar N, Bhuiyan MIH, Graham SH. Abolishing UCHL1's hydrolase activity exacerbates ischemia-induced axonal injury and functional deficits in mice. J Cereb Blood Flow Metab 2024; 44:1349-1361. [PMID: 38833565 PMCID: PMC11542126 DOI: 10.1177/0271678x241258809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/26/2024] [Accepted: 03/18/2024] [Indexed: 06/06/2024]
Abstract
Ubiquitin C-terminal hydrolase L1 (UCHL1) is a neuronal protein important in maintaining axonal integrity and motor function and may be important in the pathogenesis of many neurological disorders. UCHL1 may ameliorate acute injury and improve recovery after cerebral ischemia. In the current study, the hypothesis that UCHL1's hydrolase activity underlies its effect in maintaining axonal integrity and function is tested after ischemic injury. Hydrolase activity was inhibited by treatment with a UCHL1 hydrolase inhibitor or by employing knockin mice bearing a mutation in the hydrolase active site (C90A). Ischemic injury was induced by oxygen-glucose deprivation (OGD) in brain slice preparations and by transient middle cerebral artery occlusion (tMCAO) surgery in mice. Hydrolase activity inhibition increased restoration time and decreased the amplitude of evoked axonal responses in the corpus callosum after OGD. Mutation of the hydrolase active site exacerbated white matter injury as detected by SMI32 immunohistochemistry, and motor deficits as detected by beam balance and cylinder testing after tMCAO. These results demonstrate that UCHL1 hydrolase activity ameliorates white matter injury and functional deficits after acute ischemic injury and support the hypothesis that UCHL1 activity plays a significant role in preserving white matter integrity and recovery of function after cerebral ischemia.
Collapse
Affiliation(s)
- Zhiping Mi
- Department of Neurology, School of Medicine, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nadya Povysheva
- Department of Neuroscience, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marie E Rose
- Department of Neurology, School of Medicine, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Ma
- Department of Neurology, School of Medicine, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dennis J Zeh
- Department of Neurology, School of Medicine, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nikitha Harikumar
- Department of Neuroscience, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohammad Iqbal H Bhuiyan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, USA
| | - Steven H Graham
- Department of Neurology, School of Medicine, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Mwema A, Gratpain V, Ucakar B, Vanvarenberg K, Perdaens O, van Pesch V, Muccioli GG, des Rieux A. Impact of calcitriol and PGD 2-G-loaded lipid nanocapsules on oligodendrocyte progenitor cell differentiation and remyelination. Drug Deliv Transl Res 2024; 14:3128-3146. [PMID: 38366115 DOI: 10.1007/s13346-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 02/18/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating and inflammatory disease of the central nervous system (CNS) in need of a curative treatment. MS research has recently focused on the development of pro-remyelinating treatments and neuroprotective therapies. Here, we aimed at favoring remyelination and reducing neuro-inflammation in a cuprizone mouse model of brain demyelination using nanomedicines. We have selected lipid nanocapsules (LNC) coated with the cell-penetrating peptide transactivator of translation (TAT), loaded with either a pro-remyelinating compound, calcitriol (Cal-LNC TAT), or an anti-inflammatory bioactive lipid, prostaglandin D2-glycerol ester (PGD2-G) (PGD2-G-LNC TAT). Following the characterization of these formulations, we showed that Cal-LNC TAT in combination with PGD2-G-LNC TAT increased the mRNA expression of oligodendrocyte differentiation markers both in the CG-4 cell line and in primary mixed glial cell (MGC) cultures. However, while the combination of Cal-LNC TAT and PGD2-G-LNC TAT showed promising results in vitro, no significant impact, in terms of remyelination, astrogliosis, and microgliosis, was observed in vivo in the corpus callosum of cuprizone-treated mice following intranasal administration. Thus, although calcitriol's beneficial effects have been abundantly described in the literature in the context of MS, here, we show that the different doses of calcitriol tested had a negative impact on the mice well-being and showed no beneficial effect in the cuprizone model in terms of remyelination and neuro-inflammation, alone and when combined with PGD2-G-LNC TAT.
Collapse
Affiliation(s)
- Ariane Mwema
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Viridiane Gratpain
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Océane Perdaens
- Cellular and Molecular Division, Institute of Neuroscience, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 53, 1200, Brussels, Belgium
| | - Vincent van Pesch
- Cellular and Molecular Division, Institute of Neuroscience, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 53, 1200, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium.
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium.
| |
Collapse
|
4
|
Zhang Y, Zhao Y, Wang Y, Li J, Huang Y, Lyu F, Wang Y, Wei P, Yuan Y, Fu Y, Gao Y. Microglial histone deacetylase 2 is dispensable for functional and histological outcomes in a mouse model of traumatic brain injury. J Cereb Blood Flow Metab 2024; 44:817-835. [PMID: 38069842 PMCID: PMC11197137 DOI: 10.1177/0271678x231197173] [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: 06/20/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 04/26/2024]
Abstract
The Class-I histone deacetylases (HDACs) mediate microglial inflammation and neurological dysfunction after traumatic brain injury (TBI). However, whether the individual Class-I HDACs play an indispensable role in TBI pathogenesis remains elusive. HDAC2 has been shown to upregulate pro-inflammatory genes in myeloid cells under brain injuries such as intracerebral hemorrhage, thereby worsening outcomes. Thus, we hypothesized that HDAC2 drives microglia toward a pro-inflammatory neurotoxic phenotype in a murine model of controlled cortical impact (CCI). Our results revealed that HDAC2 expression was highly induced in CD16/CD32+ pro-inflammatory microglia 3 and 7d after TBI. Surprisingly, microglia-targeted HDAC2 knockout (HDAC2 miKO) mice failed to demonstrate a beneficial phenotype after CCI/TBI compared to their wild-type (WT) littermates. HDAC2 miKO mice exhibited comparable levels of grey and white matter injury, efferocytosis, and sensorimotor and cognitive deficits after CCI/TBI as WT mice. RNA sequencing of isolated microglia 3d after CCI/TBI indicated the elevation of a panel of pro-inflammatory cytokines/chemokines in HDAC2 miKO mice over WT mice, and flow cytometry showed further elevated brain infiltration of neutrophils and B cells in HDAC2 miKO mice. Together, this study does not support a detrimental role for HDAC2 in microglial responses after TBI and calls for investigation into alternative mechanisms.
Collapse
Affiliation(s)
- Yue Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yongfang Zhao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yana Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Fan Lyu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yangfan Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Pengju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yiwen Yuan
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yi Fu
- Department of Neurology & Institute of Neurology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Huang Y, Meng S, Wu B, Shi H, Wang Y, Xiang J, Li J, Shi Z, Wu G, Lyu Y, Jia X, Hu J, Xu ZX, Gao Y. HSPB2 facilitates neural regeneration through autophagy for sensorimotor recovery after traumatic brain injury. JCI Insight 2023; 8:e168919. [PMID: 37606039 PMCID: PMC10543718 DOI: 10.1172/jci.insight.168919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/06/2023] [Indexed: 08/23/2023] Open
Abstract
Autophagy is a promising target for promoting neural regeneration, which is essential for sensorimotor recovery following traumatic brain injury (TBI). Whether neuronal heat shock protein B2 (HSPB2), a small molecular heat shock protein, reduces injury and promotes recovery following TBI remains unclear. In this study, we demonstrated that HSPB2 was significantly increased in the neurons of a TBI mouse model, patients, and primary neuron cultures subjected to oxygen/glucose deprivation and reperfusion treatment. Upon creating a tamoxifen-induced neuron-specific HSPB2 overexpression transgenic mouse model, we found that elevated HSPB2 levels promoted long-term sensorimotor recovery and alleviated tissue loss after TBI. We also demonstrated that HSPB2 enhanced white matter structural and functional integrity, promoted central nervous system (CNS) plasticity, and accelerated long-term neural remodeling. Moreover, we found that autophagy occurred around injured brain tissues in patients, and the pro-regenerative effects of HSPB2 relied on its autophagy-promoting function. Mechanistically, HSPB2 may regulate autophagy possibly by forming the HSPB2/BCL2-associated athanogene 3/sequestosome-1 complex to facilitate the clearance of erroneously accumulated proteins in the axons. Treatment with the autophagy inhibitor chloroquine during the acute stage or delayed induction of HSPB2 remarkably impeded HSPB2's long-term reparative function, indicating the importance of acute-stage autophagy in long-term neuro-regeneration. Our findings highlight the beneficial role of HSPB2 in neuro-regeneration and functional recovery following acute CNS injury, thereby emphasizing the therapeutic potential of autophagy regulation for enhancing neuro-regeneration.
Collapse
Affiliation(s)
- Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Shan Meng
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Biwu Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong Shi
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yana Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Jiakun Xiang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Ziyu Shi
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Gang Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanchen Lyu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Xu Jia
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhi-Xiang Xu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science; Institutes of Brain Science; and
| |
Collapse
|
6
|
Bokobza C, Jacquens A, Guenoun D, Bianco B, Galland A, Pispisa M, Cruz A, Zinni M, Faivre V, Roumier A, Lebon S, Vitalis T, Csaba Z, Le Charpentier T, Schwendimann L, Young-Ten P, Degos V, Monteiro P, Dournaud P, Gressens P, Van Steenwinckel J. Targeting the brain 5-HT7 receptor to prevent hypomyelination in a rodent model of perinatal white matter injuries. J Neural Transm (Vienna) 2023; 130:281-297. [PMID: 36335540 PMCID: PMC10033587 DOI: 10.1007/s00702-022-02556-8] [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: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Approximately 15 million babies are born prematurely every year and many will face lifetime motor and/or cognitive deficits. Children born prematurely are at higher risk of developing perinatal brain lesions, especially white matter injuries (WMI). Evidence in humans and rodents demonstrates that systemic inflammation-induced neuroinflammation, including microglial and astrocyte reactivity, is the prominent processes of WMI associated with preterm birth. Thus, a new challenge in the field of perinatal brain injuries is to develop new neuroprotective strategies to target neuroinflammation to prevent WMI. Serotonin (5-HT) and its receptors play an important role in inflammation, and emerging evidence indicates that 5-HT may regulate brain inflammation by the modulation of microglial reactivity and astrocyte functions. The present study is based on a mouse model of WMI induced by intraperitoneal (i.p.) injections of IL-1β during the first 5 days of life. In this model, certain key lesions of preterm brain injuries can be summarized by (i) systemic inflammation, (ii) pro-inflammatory microglial and astrocyte activation, and (iii) inhibition of oligodendrocyte maturation, leading to hypomyelination. We demonstrate that Htr7 mRNA (coding for the HTR7/5-HT7 receptor) is significantly overexpressed in the anterior cortex of IL-1β-exposed animals, suggesting it as a potential therapeutic target. LP-211 is a specific high-affinity HTR7 agonist that crosses the blood-brain barrier (BBB). When co-injected with IL-1β, LP-211 treatment prevented glial reactivity, the down-regulation of myelin-associated proteins, and the apparition of anxiety-like phenotypes. Thus, HTR7 may represent an innovative therapeutic target to protect the developing brain from preterm brain injuries.
Collapse
Affiliation(s)
- Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France.
| | - Alice Jacquens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Anesthesia and Critical Care, APHP-Sorbonne University, Hôpital La Pitié- Salpêtrière, Paris, France
| | - David Guenoun
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Pharmacy, APHP, Hôpital Robert Debré, Université de Paris, Paris, France
| | - Blandine Bianco
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Anne Galland
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Maxime Pispisa
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Alexandra Cruz
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Manuela Zinni
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Anne Roumier
- Sorbonne Université, Inserm, UMR-S 1270, Paris, France
| | - Sophie Lebon
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Tania Vitalis
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Zsolt Csaba
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | | | | | | | - Vincent Degos
- Department of Anesthesia and Critical Care, APHP-Sorbonne University, Hôpital La Pitié- Salpêtrière, Paris, France
| | - Patricia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Pascal Dournaud
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | | |
Collapse
|
7
|
Liu L, Du X, Yang Q, Li M, Ran Q, Liu Q, Yang L, Sun L, Guo Y, Li Y, Chen Y, Zhu X, Li Q. Ginsenoside Rg1 promotes remyelination and functional recovery in demyelinating disease by enhancing oligodendrocyte precursor cells-mediated myelin repair. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154309. [PMID: 35994846 DOI: 10.1016/j.phymed.2022.154309] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Inefficient differentiation of oligodendrocyte precursor cells (OPCs) is one of the significant pathological obstacles of myelin repair and provides an essential therapeutic target against behavioral dysfunction in various neurodegenerative diseases, especially in secondary progressive multiple sclerosis (SPMS). Ginsenoside Rg1 (Rg1) has traditionally been recognized as a protector of neuronal damages, preventing its degeneration. PURPOSE We investigated the effects of Rg1 on myelin regeneration-mediated by OPCs and its therapeutic significance in SPMS. METHODS A cuprizone (CPZ) model was established and then administered with Rg1 specific for evaluations of functional recovery and remyelination. In vitro, the primary mouse OPCs were isolated and cultured for examining their ability of myelin repair. Furthermore, a chronic experimental autoimmune encephalomyelitis (EAE) model was utilized to assess the therapeutic value on SPMS. RESULTS We found that Rg1 promoted functional recovery of the demyelinated mice, including spatial memory, motor function, and anxiety-like behavior. Histologically, Rg1 enhanced myelin-genesis as proven by myelin staining and microstructures of myelin observed by transmission electron microscope. Furthermore, Rg1 significantly increased Olig2+ oligodendrocyte lineage cells in callosum, implying that the pro-remyelination effect of Rg1 was closely correlated to the enhanced differentiation of OPCs. We further demonstrated that Rg1 increased the survival and proliferation of OPCs as well as induced maturation in oligodendrocytes (OLs). Molecular analysis showed that Rg1 transduced the pro-differentiation signaling programmed by the GSK3β/β-Catenin pathway. Notably, relying on its pro-remyelination effects, Rg1 ameliorated severity and histopathology of EAE disease. CONCLUSION By paving the way for OPCs differentiation, Rg1 could maintain the integrity of myelin and is a promising candidate for functional recovery in demyelinating diseases.
Collapse
Affiliation(s)
- Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xinke Du
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Manjing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingsen Ran
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingwu Liu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Lina Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lisong Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuxuan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
8
|
Wang K, Wang C, Chen D, Huang Y, Li J, Wei P, Shi Z, Zhang Y, Gao Y. The role of microglial/macrophagic salt-inducible kinase 3 on normal and excessive phagocytosis after transient focal cerebral ischemia. Cell Mol Life Sci 2022; 79:439. [PMID: 35864266 PMCID: PMC9304053 DOI: 10.1007/s00018-022-04465-1] [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: 03/02/2022] [Revised: 06/17/2022] [Accepted: 07/03/2022] [Indexed: 11/28/2022]
Abstract
Previous studies suggested that anti-inflammatory microglia/macrophages (Mi/MΦ) play a role in “normal phagocytosis,” which promoted the rapid clearance of necrotic substances and apoptotic cells. More recently, a few studies have found that Mi/MΦ also play a role in “pathological phagocytosis” in the form of excessive or reduced phagocytosis, thereby worsening damage induced by CNS diseases. However, the underlying mechanisms and the Mi/MΦ subtypes related to this pathological phagocytosis are still unknown. Salt-inducible kinase 3 (SIK3), a member of the 5’ adenosine monophosphate-activated protein kinase (AMPK) family, has been shown to regulate inflammation in several peripheral diseases. Whether SIK3 also regulates the inflammatory response in CNS diseases is currently unknown. Therefore, in this study, we created a transgenic tamoxifen-induced Mi/MΦ-specific SIK3 conditional knockout (SIK3-cKO) mouse to examine SIK3’s role in phagocytotic function induced by transient focal cerebral ischemia (tFCI). By single-cell RNA-seq, we found the pro-inflammatory Mi/MΦ phenotype performed an excessive phagocytotic function, but the anti-inflammatory Mi/MΦ phenotype performed a normal phagocytotic function. We found that SIK3-cKO caused Mi/MΦ heterogenization from the transitional phenotype to the anti-inflammatory phenotype after tFCI. This phenotypic shift corresponded with enhanced phagocytosis of both apoptotic and live neurons. Interestingly, SIK3-cKO enhanced normal phagocytosis of myelin debris but attenuating excessive phagocytosis of non-damaged myelin sheath, thereby protecting white matter integrity after tFCI. CD16, a pro-inflammation marker, was decreased significantly by SIK3-cKO and correlated with “excessive phagocytosis.” SIK3-cKO promoted long-term recovery of white matter function and neurological function as assessed with electrophysiological compound action potential (CAPs) and behavioral analysis. This study is the first to show a role of SIK3 in Mi/MΦ phagocytosis in CNS diseases, and reveals that promoting Mi/MΦ anti-inflammatory heterogenization inhibits “excessive phagocytosis” of live cells and facilitates “normal phagocytosis” of apoptotic cells. Therefore, inhibition of SIK3 in Mi/MΦ may be a potential therapeutic target in stroke and other CNS diseases with accompanying white matter destruction.
Collapse
Affiliation(s)
- Ke Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Chenran Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Di Chen
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Pengju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ziyu Shi
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yue Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Chen D, Li J, Huang Y, Wei P, Miao W, Yang Y, Gao Y. Interleukin 13 promotes long-term recovery after ischemic stroke by inhibiting the activation of STAT3. J Neuroinflammation 2022; 19:112. [PMID: 35578342 PMCID: PMC9109418 DOI: 10.1186/s12974-022-02471-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Microglia/macrophages are activated after cerebral ischemic stroke and can contribute to either brain injury or recovery by polarizing microglia/macrophage into distinctive functional phenotypes with pro- or anti-inflammatory properties. Interleukin-13 (IL-13) is an anti-inflammatory cytokine that regulates microglia/macrophage polarization toward an anti-inflammatory phenotype. However, it is not clear whether IL-13 is beneficial after ischemic stroke long-term and the underlying molecular mechanism(s) remain unknown. Thus, we examined the effect of IL-13 on long-term recovery and microglia/macrophage polarization in mice with transient middle cerebral artery occlusion model (tMCAO). METHODS tMCAO was induced in adult male C57BL/6J mice. IL-13 (60 μg/kg) was administered intranasally starting 2 h after stroke and continued for seven consecutive days. Sensorimotor function, spatial learning and memory function, as well as brain infarct volume were assessed up to 35 days after stroke. White matter integrity was evaluated by electrophysiology, immunofluorescence staining, and transmission electron microscopy. Microglia/macrophage activation was assessed using immunofluorescence staining and quantitative real-time polymerase chain reaction. Changes in immune cells in the brain and the periphery, and expression of IL-13 receptors in different brain cells were detected by flow cytometry. Primary neuron/microglia co-cultures and a STAT3 inhibitor were used for mechanistic studies. RESULTS Post-treatment with IL-13 improved long-term neurofunctional recovery and decreased brain tissue atrophy after stroke. Intranasal delivery of IL-13 enhanced the structural and functional integrity of white matter after stroke. Furthermore, the neuroprotection afforded by IL-13 administration was not due to a direct effect on neurons, but by indirectly regulating the anti-inflammatory phenotype of microglia/macrophages. IL-13 treatment also had no effect on peripheral immune cells. Mechanistically, IL-13 improved the long-term outcome after ischemic stroke by promoting the polarization of microglia/macrophages toward the anti-inflammatory phenotype at least partially by inhibiting the phosphorylation of STAT3. CONCLUSIONS IL-13 promotes white matter repair and improves neurofunctional outcomes after ischemic stroke by modulating microglia/macrophages via inhibition of STAT3 phosphorylation.
Collapse
Affiliation(s)
- Di Chen
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Pengju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Wanying Miao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Yaomei Yang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China.
| |
Collapse
|
10
|
Sen MK, Hossain MJ. Oligodendrocyte-Specific Mechanisms of Myelin Thinning: Implications for Neurodegenerative Diseases. Front Neurosci 2021; 15:663053. [PMID: 33841096 PMCID: PMC8024530 DOI: 10.3389/fnins.2021.663053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/01/2021] [Indexed: 12/21/2022] Open
Affiliation(s)
- Monokesh K Sen
- School of Medicine, Western Sydney University, Penrith, NSW, Australia.,Peter Duncan Neuroscience Research Unit, St. Vincent's Centre for Applied Medical Research, Darlinghurst, Sydney, NSW, Australia
| | | |
Collapse
|
11
|
Mu HF, Gao XG, Li SC, Wei PJ, Zhao YF, Zhang WT, Wang Y, Gao YQ. Distinctive functional deficiencies in axonal conduction associated with two forms of cerebral white matter injury. CNS Neurosci Ther 2019; 25:1018-1029. [PMID: 31140740 PMCID: PMC6698976 DOI: 10.1111/cns.13155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/09/2019] [Accepted: 05/11/2019] [Indexed: 01/19/2023] Open
Abstract
Aims This study determines whether assessment with compound action potentials (CAPs) can distinguish two different forms of cerebral white matter injury at the functional levels. Methods A pure demyelination model was induced in C57/BL6 adult mice by dietary supplementation of cuprizone (0.2%) for 6 weeks. Callosal L‐N5‐(1‐Iminoethyl) ornithine (L‐NIO) hydrochloride (27 mg/mL) was injected into the corpus callosum (CC) to induce a focal white matter stroke (WMS), resulting in both demyelination and axonal injury. White matter integrity was assessed by performing CAP recording, electron microscopy, and immunohistological and luxol fast blue (LFB) staining. Results Immunohistological and electron microscopic analyses confirmed the induction of robust demyelination in CC with cuprizone, and mixed demyelination and axonal damage with L‐NIO. Electrophysiologically, cuprizone‐induced demyelination significantly reduced the amplitude of negative peak 1 (N1), but increased the amplitude of negative peak 2 (N2), of the CAPs compared to the sham controls. However, cuprizone did not affect the axonal conduction velocity. In contrast, the amplitude and area of both N1 and N2 along with N1 axonal conduction velocity were dramatically decreased in L‐NIO‐induced WMS. Conclusions Concertedly, parameters of the CAPs offer a novel functional assessment strategy for cerebral white matter injury in rodent models.
Collapse
Affiliation(s)
- Hong-Feng Mu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu-Guang Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Si-Cheng Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng-Ju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong-Fang Zhao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Ting Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan-Qin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Neurology Department of Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|