1
|
Zhu J, Xu N, Lin H, Deng L, Xie B, Jiang X, Liao R, Yang C. Remote ischemic preconditioning plays a neuroprotective role in cerebral ischemia-reperfusion mice by inhibiting mitophagy. Heliyon 2024; 10:e39076. [PMID: 39640619 PMCID: PMC11620096 DOI: 10.1016/j.heliyon.2024.e39076] [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: 03/14/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 12/07/2024] Open
Abstract
Remote ischemic preconditioning (RIPC) represents a clinically feasible method for safeguarding vital organs against ischemic injury. However, its specific role in cerebral ischemia-reperfusion (I/R) injury remains to be definitively elucidated. In this study, we investigated the neuroprotective effects of RIPC on mice at 7 days post-cerebral I/R and its involvement in mitophagy and mitochondrial dysfunction. Cerebral I/R led to impaired brain function, as well as structural and functional damage to mitochondria. Notably, RIPC treatment ameliorated the neurological dysfunction induced by cerebral I/R. Compared with the I/R group, the expression levels of NeuN, MBP, PDH, and Tom20 were significantly elevated in the RIPC + I/R group. Furthermore, mitochondria in the RIPC + I/R group exhibited more intact structure compared to those in the I/R group. In mice subjected to I/R injury, RIPC treatment markedly increased ATP content, ADP content, TAN level and glucose uptake while upregulating expression levels of Parkin, Pink1 and P62 proteins; it also reduced both the volume of ischemic foci and the number of mitochondrial autophagosomes along with decreasing LC3B II/I ratio. In conclusion, RIPC may exert a neuroprotective role by inhibiting excessive mitophagy during subacute stages following an ischemic stroke.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Na Xu
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Heng Lin
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqian Jiang
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Runde Liao
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chaoxian Yang
- Department of Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Michalettos G, Clausen F, Rostami E, Marklund N. Post-injury treatment with 7,8-dihydroxyflavone attenuates white matter pathology in aged mice following focal traumatic brain injury. Neurotherapeutics 2024; 22:e00472. [PMID: 39428261 DOI: 10.1016/j.neurot.2024.e00472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Traumatic brain injury (TBI) is a major cause of morbidity and mortality, not least in the elderly. The incidence of aged TBI patients has increased dramatically during the last decades. High age is a highly negative prognostic factor in TBI, and pharmacological treatment options are lacking. We used the controlled cortical impact (CCI) TBI model in 23-month-old male and female mice and analyzed the effect of post-injury treatment with 7,8 dihydroxyflavone (7,8-DHF), a brain-derived neurotrophic factor (BDNF)-mimetic compound, on white matter pathology. Following CCI or sham injury, mice received subcutaneous 7,8-DHF injections (5 mg/kg) 30 min post-injury and were sacrificed on 2, 7 or 14 days post-injury (dpi) for histological and immunofluorescence analyses. Histological assessment with Luxol Fast Blue (LFB)/Cresyl Violet stain showed that administration of 7,8-DHF resulted in preserved white matter tissue at 2 and 7 dpi with no difference in cortical tissue loss at all investigated time points. Treatment with 7,8-DHF led to reduced axonal swellings at 2 and 7 dpi, as visualized by SMI-31 (Neurofilament Heavy Chain) immunofluorescence, and reduced number of TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labelling)/CC1-positive mature oligodendrocytes at 2 dpi in the perilesional white matter. Post-injury proliferation of Platelet-derived Growth Factor Receptor (PDGFRα)-positive oligodendodrocyte progenitor cells was not altered by 7,8-DHF. Our results suggest that 7,8-DHF can attenuate white matter pathology by mitigating axonal injury and oligodendrocyte death in the aged mouse brain following TBI. These data argue that further exploration of 7,8-DHF towards clinical use is warranted.
Collapse
Affiliation(s)
- Georgios Michalettos
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Fredrik Clausen
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Elham Rostami
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University and Lund University Hospital, Lund, Sweden.
| |
Collapse
|
3
|
Gómez-de Frutos MC, Laso-García F, García-Suárez I, Piniella D, Otero-Ortega L, Alonso-López E, Pozo-Novoa J, Gallego-Ruiz R, Díaz-Gamero N, Fuentes B, Alonso de Leciñana M, Díez-Tejedor E, Ruiz-Ares G, Gutiérrez-Fernández M. The impact of experimental diabetes on intracerebral haemorrhage. A preclinical study. Biomed Pharmacother 2024; 176:116834. [PMID: 38815288 DOI: 10.1016/j.biopha.2024.116834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024] Open
Abstract
Although diabetes mellitus negatively affects post-ischaemic stroke injury and recovery, its impact on intracerebral haemorrhage (ICH) remains uncertain. This study aimed to investigate the effect of experimental diabetes (ED) on ICH-induced injury and neurological impairment. Sprague-Dawley rats were induced with ED 2 weeks before ICH induction. Animals were randomly assigned to four groups: 1)Healthy; 2)ICH; 3)ED; 4)ED-ICH. ICH and ED-ICH groups showed similar functional assessment. The ED-ICH group exhibited significantly lower haemorrhage volume compared with the ICH group, except at 1 mo. The oedema/ICH volume ratio and cistern displacement ratio were significantly higher in the ED-ICH group. Vascular markers revealed greater expression of α-SMA in the ED groups (ED and ED-ICH) compared with ICH. Conversely, the ICH groups (ED-ICH and ICH) exhibited higher levels of VEGF compared to the healthy and ED groups. An assessment of myelin tract integrity showed an increase in fractional anisotropy in the ED and ED-ICH groups compared with ICH. The ED group showed higher cryomyelin expression than the ED-ICH and ICH groups. Additionally, the ED groups (ED and ED-ICH) displayed higher expression of MOG and Olig-2 than ICH. As for inflammation, MCP-1 levels were significantly lower in the ED-ICH groups compared with the ICH group. Notably, ED did not aggravate the neurological outcome; however, it results in greater ICH-related brain oedema, greater brain structure displacement and lower haemorrhage volume. ED influences the cerebral vascularisation with an increase in vascular thickness, limits the inflammatory response and attenuates the deleterious effect of ICH on white matter integrity.
Collapse
Affiliation(s)
- Mari Carmen Gómez-de Frutos
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain; Faculty HM Hospitals of Health Sciences, Universidad Camilo José Cela, Villanueva de la Cañada, Madrid 28692, Spain
| | - Fernando Laso-García
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Iván García-Suárez
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain; Department of Emergency Service, San Agustín University Hospital, Asturias, Spain
| | - Dolores Piniella
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain; Faculty of Medicine, Universidad Alfonso X el Sabio, Villanueva de la Cañada, Madrid 28691, Spain
| | - Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Elisa Alonso-López
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Javier Pozo-Novoa
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Rebeca Gallego-Ruiz
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Nerea Díaz-Gamero
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Blanca Fuentes
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - María Alonso de Leciñana
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain
| | - Gerardo Ruiz-Ares
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain.
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area, Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital, Universidad Autónoma de Madrid), Madrid, Spain.
| |
Collapse
|
4
|
Daca A, Jarzembowski T. From the Friend to the Foe- Enterococcus faecalis Diverse Impact on the Human Immune System. Int J Mol Sci 2024; 25:2422. [PMID: 38397099 PMCID: PMC10888668 DOI: 10.3390/ijms25042422] [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: 01/30/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Enterococcus faecalis is a bacterium which accompanies us from the first days of our life. As a commensal it produces vitamins, metabolizes nutrients, and maintains intestinal pH. All of that happens in exchange for a niche to inhabit. It is not surprising then, that the bacterium was and is used as an element of many probiotics and its positive impact on the human immune system and the body in general is hard to ignore. This bacterium has also a dark side though. The plasticity and relative ease with which one acquires virulence traits, and the ability to hide from or even deceive and use the immune system to spread throughout the body make E. faecalis a more and more dangerous opponent. The statistics clearly show its increasing role, especially in the case of nosocomial infections. Here we present the summarization of current knowledge about E. faecalis, especially in the context of its relations with the human immune system.
Collapse
Affiliation(s)
- Agnieszka Daca
- Department of Physiopathology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Tomasz Jarzembowski
- Department of Microbiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
5
|
Treble-Barna A, Petersen BA, Stec Z, Conley YP, Fink EL, Kochanek PM. Brain-Derived Neurotrophic Factor in Pediatric Acquired Brain Injury and Recovery. Biomolecules 2024; 14:191. [PMID: 38397427 PMCID: PMC10886547 DOI: 10.3390/biom14020191] [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: 12/20/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
We review emerging preclinical and clinical evidence regarding brain-derived neurotrophic factor (BDNF) protein, genotype, and DNA methylation (DNAm) as biomarkers of outcomes in three important etiologies of pediatric acquired brain injury (ABI), traumatic brain injury, global cerebral ischemia, and stroke. We also summarize evidence suggesting that BDNF is (1) involved in the biological embedding of the psychosocial environment, (2) responsive to rehabilitative therapies, and (3) potentially modifiable. BDNF's unique potential as a biomarker of neuroplasticity and neural repair that is reflective of and responsive to both pre- and post-injury environmental influences separates it from traditional protein biomarkers of structural brain injury with exciting potential to advance pediatric ABI management by increasing the accuracy of prognostic tools and informing clinical decision making through the monitoring of therapeutic effects.
Collapse
Affiliation(s)
- Amery Treble-Barna
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Bailey A. Petersen
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Zachary Stec
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
| | - Yvette P. Conley
- Department of Health Promotion & Development, University of Pittsburgh School of Nursing, Pittsburgh, PA 15213, USA;
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Mirzahosseini G, Adam JM, Nasoohi S, El-Remessy AB, Ishrat T. Lost in Translation: Neurotrophins Biology and Function in the Neurovascular Unit. Neuroscientist 2023; 29:694-714. [PMID: 35769016 DOI: 10.1177/10738584221104982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The neurovascular unit (NVU) refers to the functional building unit of the brain and the retina, where neurons, glia, and microvasculature orchestrate to meet the demand of the retina's and brain's function. Neurotrophins (NTs) are structural families of secreted proteins and are known for exerting neurotrophic effects on neuronal differentiation, survival, neurite outgrowth, synaptic formation, and plasticity. NTs include several molecules, such as nerve growth factor, brain-derived neurotrophic factor, NT-3, NT-4, and their precursors. Furthermore, NTs are involved in signaling pathways such as inflammation, apoptosis, and angiogenesis in a nonneuronal cell type. Interestingly, NTs and the precursors can bind and activate the p75 neurotrophin receptor (p75NTR) at low and high affinity. Mature NTs bind their cognate tropomyosin/tyrosine-regulated kinase receptors, crucial for maintenance and neuronal development in the brain and retina axis. Activation of p75NTR results in neuronal apoptosis and cell death, while tropomysin receptor kinase upregulation contributes to differentiation and cell growth. Recent findings indicate that modulation of NTs and their receptors contribute to neurovascular dysfunction in the NVU. Several chronic metabolic and acute ischemic diseases affect the NVU, including diabetic and ischemic retinopathy for the retina, as well as stroke, acute encephalitis, and traumatic brain injury for the brain. This work aims to review the current evidence through published literature studying the impact of NTs and their receptors, including the p75NTR receptor, on the injured and healthy brain-retina axis.
Collapse
Affiliation(s)
- Golnoush Mirzahosseini
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Justin Mark Adam
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
7
|
Wang Y, Jiang A, Yan J, Wen D, Gu N, Li Z, Sun X, Wu Y, Guo Z. Inhibition of GPR17/ID2 Axis Improve Remyelination and Cognitive Recovery after SAH by Mediating OPC Differentiation in Rat Model. Transl Stroke Res 2023:10.1007/s12975-023-01201-0. [PMID: 37935878 DOI: 10.1007/s12975-023-01201-0] [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: 08/07/2023] [Revised: 09/21/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023]
Abstract
Myelin sheath injury contributes to cognitive deficits following subarachnoid hemorrhage (SAH). G protein-coupled receptor 17 (GPR17), a membrane receptor, negatively regulates oligodendrocyte precursor cell (OPC) differentiation in both developmental and pathological contexts. Nonetheless, GPR17's role in modulating OPC differentiation, facilitating remyelination post SAH, and its interaction with downstream molecules remain elusive. In a rat SAH model induced by arterial puncture, OPCs expressing GPR17 proliferated prominently by day 14 post-onset, coinciding with compromised myelin sheath integrity and cognitive deficits. Selective Gpr17 knockdown in oligodendrocytes (OLs) via adeno-associated virus (AAV) administration revealed that reduced GPR17 levels promoted OPC differentiation, restored myelin sheath integrity, and improved cognitive deficits by day 14 post-SAH. Moreover, GPR17 knockdown attenuated the elevated expression of the inhibitor of DNA binding 2 (ID2) post-SAH, suggesting a GPR17-ID2 regulatory axis. Bi-directional modulation of ID2 expression in OLs using AAV unveiled that elevated ID2 counteracted the restorative effects of GPR17 knockdown. This resulted in hindered differentiation, exacerbated myelin sheath impairment, and worsened cognitive deficits. These findings highlight the pivotal roles of GPR17 and ID2 in governing OPC differentiation and axonal remyelination post-SAH. This study positions GPR17 as a potential therapeutic target for SAH intervention. The interplay between GPR17 and ID2 introduces a novel avenue for ameliorating cognitive deficits post-SAH.
Collapse
Affiliation(s)
- Yingwen Wang
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Anan Jiang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Yan
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Daochen Wen
- Department of Neurosurgery, Xuanhan County People's Hospital, Dazhou, China
| | - Nina Gu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Zhao Li
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China.
| | - Zongduo Guo
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
8
|
Seo JS, Lee SH, Won HS, Yang M, Nahm SS, Nam SM. Effects of Gestational and Lactational Lead Exposure and High Fat Diet Feeding on Cerebellar Development of Postnatal Rat Offspring. Nutrients 2023; 15:4325. [PMID: 37892401 PMCID: PMC10609260 DOI: 10.3390/nu15204325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Obesity and heavy metals, such as lead (Pb), are detrimental to the adult brain because they impair cognitive function and structural plasticity. However, the effects of co-administration of Pb and a high-fat diet (HFD) on the developing cerebellum is not clearly elucidated. We investigated the effects of Pb exposure (0.3% lead acetate) on developing cerebellum in the pups of an HFD-fed obese rat model. One week before mating, we fed a chow diet (CD) or HFD to the rats for one week and additionally administered Pb to HFD-fed female SD rats. Thereafter, treatment with Pb and a HFD was continued during the gestational and lactational periods. On postnatal day 21, the pups were euthanized to sample the brain tissue and blood for further analysis. Blood Pb levels were significantly higher in HFD-fed rats than in CD-fed rats. Histologically, the prominent degeneration of Purkinje cells was induced by the co-administration of Pb and HFD. The calbindin-28Kd-, GAD67-, NMDAR1-, and PSD95-immunopositive Purkinje cells and inhibitory synapse-forming pinceau structures were significantly decreased following Pb and HFD co-administration. MBP-immunoreactive myelinated axonal fibers were also impaired by HFD but were significantly damaged by the co-administration of HFD and Pb. Oxidative stress-related Nrf2-HO1 signaling was activated by HFD feeding, and Pb exposure further aggravated oxidative stress, as demonstrated by the consumption of endogenous anti-oxidant in HFD-Pb rats. The pro-inflammatory response was also increased by the co-administration of HFD and Pb in the cerebellum of the rat offspring. The present results suggest that HFD and Pb treatment during the gestational and lactational periods are harmful to cerebellar development.
Collapse
Affiliation(s)
- Jin Seok Seo
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (J.S.S.); (S.-S.N.)
| | - Shin Hyo Lee
- Department of Anatomy, School of Medicine and Jesaeng-Euise Clinical Anatomy Center, Wonkwang University, Iksan 54538, Republic of Korea; (S.H.L.); (H.-S.W.); (M.Y.)
| | - Hyung-Sun Won
- Department of Anatomy, School of Medicine and Jesaeng-Euise Clinical Anatomy Center, Wonkwang University, Iksan 54538, Republic of Korea; (S.H.L.); (H.-S.W.); (M.Y.)
| | - Miyoung Yang
- Department of Anatomy, School of Medicine and Jesaeng-Euise Clinical Anatomy Center, Wonkwang University, Iksan 54538, Republic of Korea; (S.H.L.); (H.-S.W.); (M.Y.)
| | - Sang-Seop Nahm
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (J.S.S.); (S.-S.N.)
| | - Sung Min Nam
- Department of Anatomy, School of Medicine and Jesaeng-Euise Clinical Anatomy Center, Wonkwang University, Iksan 54538, Republic of Korea; (S.H.L.); (H.-S.W.); (M.Y.)
| |
Collapse
|
9
|
Ahmed W, Kuniyan MS, Jawed AM, Chen L. Engineered Extracellular Vesicles for Drug Delivery in Therapy of Stroke. Pharmaceutics 2023; 15:2173. [PMID: 37765144 PMCID: PMC10537154 DOI: 10.3390/pharmaceutics15092173] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are promising therapeutic modalities for treating neurological conditions. EVs facilitate intercellular communication among brain cells under normal and abnormal physiological conditions. The potential capability of EVs to pass through the blood-brain barrier (BBB) makes them highly promising as nanocarrier contenders for managing stroke. EVs possess several potential advantages compared to existing drug-delivery vehicles. These advantages include their capacity to surpass natural barriers, target specific cells, and stability within the circulatory system. This review explores the trafficking and cellular uptake of EVs and evaluates recent findings in the field of EVs research. Additionally, an overview is provided of the techniques researchers utilize to bioengineer EVs for stroke therapy, new results on EV-BBB interactions, and the limitations and prospects of clinically using EVs for brain therapies. The primary objective of this study is to provide a comprehensive analysis of the advantages and challenges related to engineered EVs drug delivery, specifically focusing on their application in the treatment of stroke.
Collapse
Affiliation(s)
- Waqas Ahmed
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, China;
- School of Medicine, Southeast University, Nanjing 210009, China; (M.S.K.); (A.M.J.)
| | | | - Aqil Mohammad Jawed
- School of Medicine, Southeast University, Nanjing 210009, China; (M.S.K.); (A.M.J.)
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, China;
| |
Collapse
|
10
|
Du M, Wang N, Xin X, Yan CL, Gu Y, Wang L, Shen Y. Endothelin-1–Endothelin receptor B complex contributes to oligodendrocyte differentiation and myelin deficits during preterm white matter injury. Front Cell Dev Biol 2023; 11:1163400. [PMID: 37009471 PMCID: PMC10063893 DOI: 10.3389/fcell.2023.1163400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Preterm cerebral white matter injury (WMI), a major form of prenatal brain injury, may potentially be treated by oligodendrocyte (OL) precursor cell (OPC) transplantation. However, the defective differentiation of OPCs during WMI seriously hampers the clinical application of OPC transplantation. Thus, improving the ability of transplanted OPCs to differentiate is critical to OPC transplantation therapy for WMI. We established a hypoxia–ischemia-induced preterm WMI model in mice and screened the molecules affected by WMI using single-cell RNA sequencing. We revealed that endothelin (ET)-1 and endothelin receptor B (ETB) are a pair of signaling molecules responsible for the interaction between neurons and OPCs and that preterm WMI led to an increase in the number of ETB-positive OPCs and premyelinating OLs. Furthermore, the maturation of OLs was reduced by knocking out ETB but promoted by stimulating ET-1/ETB signaling. Our research reveals a new signaling module for neuron–OPC interaction and provides new insight for therapy targeting preterm WMI.
Collapse
Affiliation(s)
- Mengjie Du
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolong Xin
- NHC and CAMS Key Laboratory of Medical Neurobiology, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chun-Lan Yan
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Gu
- Department of Stem Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Liang Wang, ; Ying Shen,
| | - Ying Shen
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Liang Wang, ; Ying Shen,
| |
Collapse
|
11
|
Kalinichenko SG, Pushchin II, Matveeva NY. Neurotoxic and cytoprotective mechanisms in the ischemic neocortex. J Chem Neuroanat 2023; 128:102230. [PMID: 36603664 DOI: 10.1016/j.jchemneu.2022.102230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Neuronal damage in ischemic stroke occurs due to permanent imbalance between the metabolic needs of the brain and the ability of the blood-vascular system to maintain glucose delivery and adequate gas exchange. Oxidative stress and excitotoxicity trigger complex processes of neuroinflammation, necrosis, and apoptosis of both neurons and glial cells. This review summarizes data on the structural and chemical changes in the neocortex and main cytoprotective effects induced by focal ischemic stroke. We focus on the expression of neurotrophins (NT) and molecular and cellular changes in neurovascular units in ischemic brain. We also discuss how these factors affect the apoptosis of cortical cells. Ischemic damage involves close interaction of a wide range of signaling molecules, each acting as an efficient marker of cell state in both the ischemic core and penumbra. NTs play the main regulatory role in brain tissue recovery after ischemic injury. Heterogeneous distribution of the BDNF, NT-3, and GDNF immunoreactivity is concordant with the selective response of different types of cortical neurons and glia to ischemic injury and allows mapping the position of viable neurons. Astrocytes are the central link in neurovascular coupling in ischemic brain by providing other cells with a wide range of vasotropic factors. The NT expression coincides with the distribution of reactive astrocytes, marking the boundaries of the penumbra. The development of ischemic stroke is accompanied by a dramatic change in the distribution of GDNF reactivity. In early ischemic period, it is mainly observed in cortical neurons, while in late one, the bulk of GDNF-positive cells are various types of glia, in particular, astrocytes. The proportion of GDNF-positive astrocytes increases gradually throughout the ischemic period. Some factors that exert cytoprotective effects in early ischemic period may display neurotoxic and pro-apoptotic effects later on. The number of apoptotic cells in the ischemic brain tissue correlates with the BDNF levels, corroborating its protective effects. Cytoprotection and neuroplasticity are two lines of brain protection and recovery after ischemic stroke. NTs can be considered an important link in these processes. To develop efficient pharmacological therapy for ischemic brain injury, we have to deepen our understanding of neurochemical adaptation of brain tissue to acute stroke.
Collapse
Affiliation(s)
- Sergei G Kalinichenko
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| | - Igor I Pushchin
- Laboratory of Physiology, A.V. Zhirmusky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| | - Natalya Yu Matveeva
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| |
Collapse
|
12
|
Sadek MA, Kandil EA, El Sayed NS, Sayed HM, Rabie MA. Semaglutide, a novel glucagon-like peptide-1 agonist, amends experimental autoimmune encephalomyelitis-induced multiple sclerosis in mice: Involvement of the PI3K/Akt/GSK-3β pathway. Int Immunopharmacol 2023; 115:109647. [PMID: 36584570 DOI: 10.1016/j.intimp.2022.109647] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/17/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is a disabling neurodegenerative disease that causes demyelination and axonal degeneration of the central nervous system. Current treatments are partially effective in managing MS relapses and have a negligible impact on treating MS cognitive deficits and cannot enhance neuronal remyelination, imposing a need for a new MS remedy. Semaglutide, a novel glucagon-like peptide-1 agonist, has recently displayed a neuroprotective effect on several neurodegenerative diseases, suggesting that it may have a protective effect in MS. Therefore, this study was conducted to investigate the influence of semaglutide on experimental autoimmune encephalomyelitis (EAE)-induced MS in mice. Here, EAE was induced in mice using spinal cord homogenate, which eventually altered the mice's cognitive and motor functions, similar to what is observed in MS. Interestingly, intraperitoneally administered semaglutide (25 nmol/kg/day) amended EAE-induced cognitive and motor deficits observed in novel object recognition, open field, rotarod, and grip strength tests. Moreover, histological examination revealed that semaglutide treatment attenuated hippocampal damage and corpus callosum demyelination caused by EAE. Additionally, biochemical testing revealed that semaglutide activates the PI3K/Akt axis, which eventually hampers GSK-3β activity. GSK-3β activity inhibition attenuates demyelination and triggers remyelination through CREB/BDNF; furthermore, it boosts Nrf2 and SOD levels, protecting the mice from EAE-induced oxidative stress. Additionally, GSK-3β inhibition minimizes neuroinflammation, as reflected by decreased NF-kβ and TNF-α levels. In conclusion, semaglutide has a neuroprotective effect in EAE-induced MS in mice, which is mediated by activating the ramified PI3K/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Helmy M Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
13
|
ZHOU ZA, WANG SR, ZHANG J, GUO RH, GUO B. Progress on acupuncture in relieving stroke induced limb spasticity by regulating neuroplasticity-related signals 针刺通过调节脑可塑性相关信号缓解脑卒中肢体痉挛状态研究进展. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2023. [DOI: 10.1016/j.wjam.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
14
|
Beresewicz-Haller M. Hippocampal region-specific endogenous neuroprotection as an approach in the search for new neuroprotective strategies in ischemic stroke. Fiction or fact? Neurochem Int 2023; 162:105455. [PMID: 36410452 DOI: 10.1016/j.neuint.2022.105455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Ischemic stroke is the leading cause of death and long-term disability worldwide, and, while considerable progress has been made in understanding its pathophysiology, the lack of effective treatments remains a major concern. In that context, receiving more and more consideration as a promising therapeutic method is the activation of natural adaptive mechanisms (endogenous neuroprotection) - an approach that seeks to enhance and/or stimulate the endogenous processes of plasticity and protection of the neuronal system that trigger the brain's intrinsic capacity for self-defence. Ischemic preconditioning is a classic example of endogenous neuroprotection, being the process by which one or more brief, non-damaging episodes of ischemia-reperfusion (I/R) induce tissue resistance to subsequent prolonged, damaging ischemia. Another less-known example is resistance to an I/R episode mounted by the hippocampal region consisting of CA2, CA3, CA4 and the dentate gyrus (here abbreviated to CA2-4, DG). This can be contrasted with the ischemia-vulnerable CA1 region. There is not yet a good understanding of these different sensitivities of the hippocampal regions, and hence of the endogenous neuroprotection characteristic of CA2-4, DG. However, this region is widely reported to have properties distinct from CA1, and capable of generating resistance to an I/R episode. These include activation of neurotrophic and neuroprotective factors, greater activation of anti-excitotoxic and anti-oxidant mechanisms, increased plasticity potential, a greater energy reserve and improved mitochondrial function. This review seeks to summarize properties of CA2-4, DG in the context of endogenous neuroprotection, and then to assess the potential utility of these properties to therapeutic approaches. In so doing, it appears to represent the first such addressing of the issue of ischemia resistance attributable to CA2-4, DG.
Collapse
|
15
|
Wang Q, Diao S, Qiu H, Gao R, Wang M, Chen Q, Xiao M, Li Z, Chen C. Galectin-3 administration drives remyelination after hypoxic-ischemic induced perinatal white matter injury. Front Cell Neurosci 2022; 16:976002. [PMID: 36204450 PMCID: PMC9532057 DOI: 10.3389/fncel.2022.976002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Hypoxic-ischemic (HI) induced perinatal white matter injury (PWMI) is a major cause of neurologic disabilities characterized by selective oligodendroglial death and myelin disruption. Galectin-3 (Gal-3) modulates postnatal subventricular zone gliogenesis and attenuates ischemic injury. However, the association between Gal-3 and myelin formation still remains unclear. In this study, we first perform Gal-3 knockdown (KD) to identify the importance of Gal-3 on myelin formation. Our results show impeded myelin formation, manifested by Olig2/CC1 (+) mature oligodendrocytes number, expression of oligodendroglial maturation-associated markers (MBP and CNPase), and myelin thickness and integrity. Then we perform recombinant Gal-3 (rGal-3) administration by intracerebroventricular injection. Notably, although rGal-3 administration shows no beneficial effect on oligodendrogenesis and myelin formation under normal condition, our results show that rGal-3 administration attenuates cognitive deficits and drives remyelination after PWMI, which are coupled to signs of enhanced myelin resiliency and cognition. Also, our results indicates that the significant increases in substrates for remyelination of rGal-3 administration are accompanied by enhanced Iba-1 (microglia marker)/ Mrc1 (M2 marker) (+) microglia and decreased Iba-1/ iNOS (M1 marker) (+) microglia. Altogether, our data in this research confirm the association between Gal-3 and myelin formation, underscore its position for the capacity for remyelination and restoration of function, and unveils the efficacy of rGal-3 administration with anti-inflammatory phenotype microglia (M2 microglia) activation. Thus, the findings suggest that Gal-3 plays a significant role in myelin formation and remyelination restoration.
Collapse
Affiliation(s)
- Qian Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
- Department of Neonatology, Women and Children's Medical Center of Guangzhou, Guangzhou, China
| | - Sihao Diao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
| | - Han Qiu
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
| | - Ruiwei Gao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
| | - Minjie Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
| | - Qiufan Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
- Department of Neonatology, Women and Children's Medical Center of Guangzhou, Guangzhou, China
| | - Mili Xiao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
| | - Zhihua Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, National Health Commission, Shanghai, China
- *Correspondence: Chao Chen
| |
Collapse
|
16
|
Aabedi AA, Young JS, Chang EF, Berger MS, Hervey-Jumper SL. Involvement of White Matter Language Tracts in Glioma: Clinical Implications, Operative Management, and Functional Recovery After Injury. Front Neurosci 2022; 16:932478. [PMID: 35898410 PMCID: PMC9309688 DOI: 10.3389/fnins.2022.932478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
To achieve optimal survival and quality of life outcomes in patients with glioma, the extent of tumor resection must be maximized without causing injury to eloquent structures. Preservation of language function is of particular importance to patients and requires careful mapping to reveal the locations of cortical language hubs and their structural and functional connections. Within this language network, accurate mapping of eloquent white matter tracts is critical, given the high risk of permanent neurological impairment if they are injured during surgery. In this review, we start by describing the clinical implications of gliomas involving white matter language tracts. Next, we highlight the advantages and limitations of methods commonly used to identify these tracts during surgery including structural imaging techniques, functional imaging, non-invasive stimulation, and finally, awake craniotomy. We provide a rationale for combining these complementary techniques as part of a multimodal mapping paradigm to optimize postoperative language outcomes. Next, we review local and long-range adaptations that take place as the language network undergoes remodeling after tumor growth and surgical resection. We discuss the probable cellular mechanisms underlying this plasticity with emphasis on the white matter, which until recently was thought to have a limited role in adults. Finally, we provide an overview of emerging developments in targeting the glioma-neuronal network interface to achieve better disease control and promote recovery after injury.
Collapse
Affiliation(s)
| | | | | | | | - Shawn L. Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
17
|
Topography of neurotrophins in the rat neocortex and their role in neuron apoptosis after experimental ischemic stroke. J Chem Neuroanat 2022; 124:102122. [PMID: 35718293 DOI: 10.1016/j.jchemneu.2022.102122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022]
Abstract
Neuronal loss due to apoptosis after ischemic injury depends on the trophic support of neurons and cytoprotective effects of neurotrophins (NTs). Different NTs may activate both pro- and antiapoptotic factors. Their distribution in the ischemic core (IC) and penumbra (IP) has been poorly studied. The available data on the localization of NTs in the ischemic brain are contradictory and depend to a certain degree on the pathogenetic model used. The distribution of NTs in different layers of the ischemic cortex is also largely unknown hindering our understanding of their exact effects and targets in different zones of the ischemic brain. We examined the immunolocalization of brain-derived neurotrophic factor (BDNF), neurotrophin-3, and glial cell line-derived neurotrophic factor (GDNF) in the parietal cortex using a rat model of ischemic stroke due to permanent occlusion of the middle cerebral artery. The spatial density of immunoreactive (IR) cells varied across the cortical layers and changed with time after ischemic injury. Their distribution in the IC differed considerably from that in the IP. The immunolocalization of neurotrophins in the contralateral hemisphere was similar to that in IP. We also studied the distribution of pro- and anti-apoptotic factors in IC and IP with and without intravenous BDNF administration. In the model without BDNF administration, the proportions of Bcl-2-, p53-, caspase-3-, and Mdm2-IR cells showed different dynamics during the ischemic period. In the model with BDNF administration, Mdm2 immunoreactivity was mainly observed in pyramidal cells of layers V/VI, and Bcl-2, in interneurons of layers II and III. The dynamics of p53 immunoreactivity was opposite to that of caspase-3 throughout the ischemic period. The present results suggest that after ischemic injury, 1) the number of neurotrophin-positive cells increases in the early ischemic period and decreases afterwards; 2) there is a close metabolic relationship between astrocytes and neurons contributing to their adaptation to ischemic conditions; 3) the IP borders undergo constant changes; 4) in the IP, neuronal loss occurs mainly by apoptotic pathway throughout the ischemic period; 5) BDNF may enhance considerably antiapoptotic mechanisms with a predominance of Mdm-2 activity in pyramidal neurons.
Collapse
|
18
|
Brichko R, Soldan A, Zhu Y, Wang MC, Faria A, Albert M, Pettigrew C. Age-Dependent Association Between Cognitive Reserve Proxy and Longitudinal White Matter Microstructure in Older Adults. Front Psychol 2022; 13:859826. [PMID: 35756247 PMCID: PMC9226781 DOI: 10.3389/fpsyg.2022.859826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/12/2022] [Indexed: 01/26/2023] Open
Abstract
Objective This study examined the association of lifetime experiences, measured by a cognitive reserve (CR) composite score composed of years of education, literacy, and vocabulary measures, to level and rate of change in white matter microstructure, as assessed by diffusion tensor imaging (DTI) measures. We also examined whether the relationship between the proxy CR composite score and white matter microstructure was modified by participant age, APOE-ε4 genetic status, and level of vascular risk. Methods A sample of 192 non-demented (n = 166 cognitively normal, n = 26 mild cognitive impairment) older adults [mean age = 70.17 (SD = 8.5) years] from the BIOCARD study underwent longitudinal DTI (mean follow-up = 2.5 years, max = 4.7 years). White matter microstructure was quantified by fractional anisotropy (FA) and radial diffusivity (RD) values in global white matter tracts and medial temporal lobe (MTL) white matter tracts. Results Using longitudinal linear mixed effect models, we found that FA decreased over time and RD increased over time in both the global and MTL DTI composites, but the rate of change in these DTI measures was not related to level of CR. However, there were significant interactions between the CR composite score and age for global RD in the full sample, and for global FA, global RD, and MTL RD among those with normal cognition. These interactions indicated that among participants with a lower baseline age, higher CR composite scores were associated with higher FA and lower RD values, while among participants with higher age at baseline, higher CR composite scores were associated with lower FA and higher RD values. Furthermore, these relationships were not modified by APOE-ε4 genotype or level of vascular risk. Conclusion The association between level of CR and DTI measures differs by age, suggesting a possible neuroprotective effect of CR among late middle-aged adults that shifts to a compensatory effect among older adults.
Collapse
Affiliation(s)
- Rostislav Brichko
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Anja Soldan
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yuxin Zhu
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Mei-Cheng Wang
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Andreia Faria
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Marilyn Albert
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States,*Correspondence: Corinne Pettigrew,
| | | |
Collapse
|
19
|
Neurotrophin Signaling Impairment by Viral Infections in the Central Nervous System. Int J Mol Sci 2022; 23:ijms23105817. [PMID: 35628626 PMCID: PMC9146244 DOI: 10.3390/ijms23105817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neurotrophins, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin 3 (NT-3), NT-4, and NT-5, are proteins involved in several important functions of the central nervous system. The activation of the signaling pathways of these neurotrophins, or even by their immature form, pro-neurotrophins, starts with their recognition by cellular receptors, such as tropomyosin receptor kinase (Trk) and 75 kD NT receptors (p75NTR). The Trk receptor is considered to have a high affinity for attachment to specific neurotrophins, while the p75NTR receptor has less affinity for attachment with neurotrophins. The correct functioning of these signaling pathways contributes to proper brain development, neuronal survival, and synaptic plasticity. Unbalanced levels of neurotrophins and pro-neurotrophins have been associated with neurological disorders, illustrating the importance of these molecules in the central nervous system. Furthermore, reports have indicated that viruses can alter the normal levels of neurotrophins by interfering with their signaling pathways. This work discusses the importance of neurotrophins in the central nervous system, their signaling pathways, and how viruses can affect them.
Collapse
|
20
|
Antidepressant effects of Enterococcus faecalis 2001 through the regulation of prefrontal cortical myelination via the enhancement of CREB/BDNF and NF-κB p65/LIF/STAT3 pathways in olfactory bulbectomized mice. J Psychiatr Res 2022; 148:137-148. [PMID: 35123326 DOI: 10.1016/j.jpsychires.2022.01.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/14/2022]
Abstract
A therapeutic strategy through the gut-brain axis has been proven to be effective in treatment for depression. In our previous study, we demonstrated that Enterococcus faecalis 2001 (EF-2001) prevents colitis-induced depressive-like behavior through the gut-brain axis in mice. More recently, we found that demyelination in the prefrontal cortex (PFC) was associated with depressive-like behavior in an animal model of major depressive disorder, olfactory bulbectomized (OBX) mice. The present study investigated the effects of EF-2001 on depressive-like behaviors in OBX mice and the underlying molecular mechanisms from the perspective of myelination in the PFC. OBX mice exhibited depressive-like behaviors in the tail-suspension, splash, and sucrose preference tests, and decreased myelin and paranodal proteins along with mature oligodendrocytes in the PFC. These behavioral and biochemical changes were all prevented by treatment with EF-2001. Further, EF-2001 treatment increased brain-derived neurotrophic factor (BDNF) and leukemia inhibitory factor (LIF) in the PFC. Interestingly, an immunohistochemical analysis revealed enhanced phospho (p) -cAMP-responsive element binding protein (CREB) expression in neurons, p-nuclear factor-kappa B (NFκB) p65 (Ser536) expression in astrocytes, and p-signal transducer and activator of transcription 3 (STAT3) (Ty705) expression in mature oligodendrocytes in the PFC of OBX mice. From these results, we suggest that EF-2001 administration prevents depressive-like behaviors by regulating prefrontal cortical myelination via the enhancement of CREB/BDNF and NFκB p65/LIF/STAT3 pathways. Our findings strongly support the idea that a therapeutic strategy involving the gut microbiota may be a promising alternative treatment for alleviating symptoms of depression.
Collapse
|
21
|
Jin R, Wang M, Zhong W, Kissinger CR, Villafranca JE, Li G. J147 Reduces tPA-Induced Brain Hemorrhage in Acute Experimental Stroke in Rats. Front Neurol 2022; 13:821082. [PMID: 35309561 PMCID: PMC8925862 DOI: 10.3389/fneur.2022.821082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background and purpose J147, a novel neurotrophic compound, was originally developed to treat aging-associated neurological diseases. Based on the broad spectrum of cytoprotective effects exhibited by this compound, we investigated whether J147 has cerebroprotection for acute ischemic stroke and whether it can enhance the effectiveness of thrombolytic therapy with tissue plasminogen activator (tPA). Methods Rats were subjected to transient occlusion of the middle cerebral artery (tMCAO) by insertion of an intraluminal suture or embolic middle cerebral artery occlusion (eMCAO), and treated intravenously with J147 alone or in combination with tPA. Results We found that J147 treatment significantly reduced infarct volume when administered at 2 h after stroke onset in the tMCAO model, but had no effect in eMCAO without tPA. However, combination treatment with J147 plus tPA at 4 h after stroke onset significantly reduced infarct volume and neurological deficits at 72 h after stroke compared with saline or tPA alone groups in the eMCAO model. Importantly, the combination treatment significantly reduced delayed tPA-associated brain hemorrhage and secondary microvascular thrombosis. These protective effects were associated with J147-mediated inhibition of matrix metalloproteinase-9 (MMP9), 15-lipoxygenase-1, and plasminogen activator inhibitor (PAI) expression in the ischemic hemispheres (predominantly in ischemic cerebral endothelium). Moreover, the combination treatment significantly reduced circulating platelet activation and platelet-leukocyte aggregation compared with saline or tPA alone groups at 24 h after stroke, which might also contribute to reduced microvascular thrombosis and neuroinflammation (as demonstrated by reduced neutrophil brain infiltration and microglial activation). Conclusion Our results demonstrate that J147 treatment alone exerts cerebral cytoprotective effects in a suture model of acute ischemic stroke, while in an embolic stroke model co-administration of J147 with tPA reduces delayed tPA-induced intracerebral hemorrhage and confers cerebroprotection. These findings suggest that J147-tPA combination therapy could be a promising approach to improving the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Rong Jin
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
| | - Min Wang
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
| | - Wei Zhong
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
| | | | | | - Guohong Li
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
22
|
Yoon H, Triplet EM, Simon WL, Choi CI, Kleppe LS, De Vita E, Miller AK, Scarisbrick IA. Blocking Kallikrein 6 promotes developmental myelination. Glia 2022; 70:430-450. [PMID: 34626143 PMCID: PMC8732303 DOI: 10.1002/glia.24100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/12/2022]
Abstract
Kallikrein related peptidase 6 (Klk6) is a secreted serine protease highly expressed in oligodendrocytes and implicated in demyelinating conditions. To gain insights into the significance of Klk6 to oligodendrocyte biology, we investigated the impact of global Klk6 gene knockout on CNS developmental myelination using the spinal cord of male and female mice as a model. Results demonstrate that constitutive loss of Klk6 expression accelerates oligodendrocyte differentiation developmentally, including increases in the expression of myelin proteins such as MBP, PLP and CNPase, in the number of CC-1+ mature oligodendrocytes, and myelin thickness by the end of the first postnatal week. Co-ordinate elevations in the pro-myelinating signaling pathways ERK and AKT, expression of fatty acid 2-hydroxylase, and myelin regulatory transcription factor were also observed in the spinal cord of 7d Klk6 knockouts. LC/MS/MS quantification of spinal cord lipids showed sphingosine and sphingomyelins to be elevated in Klk6 knockouts at the peak of myelination. Oligodendrocyte progenitor cells (OPCs)-derived from Klk6 knockouts, or wild type OPCs-treated with a Klk6 inhibitor (DFKZ-251), also showed increased MBP and PLP. Moreover, inhibition of Klk6 in OPC cultures enhanced brain derived neurotrophic factor-driven differentiation. Altogether, these findings suggest that oligodendrocyte-derived Klk6 may operate as an autocrine or paracrine rheostat, or brake, on pro-myelinating signaling serving to regulate myelin homeostasis developmentally and in the adult. These findings document for the first time that inhibition of Klk6 globally, or specifically in oligodendrocyte progenitors, is a strategy to increase early stages of oligodendrocyte differentiation and myelin production in the CNS.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Erin M. Triplet
- Regenerative Sciences Program, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Whitney L. Simon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Chan-Il Choi
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Laurel S. Kleppe
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Elena De Vita
- University of Heidelberg, Faculty of Biosciences, 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Aubry K. Miller
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
- Regenerative Sciences Program, Mayo Clinic School of Biomedical Sciences Rochester 55905
- Department of Physiology and Biomedical Engineering, Minnesota USA 55905
| |
Collapse
|
23
|
Zhao D, Zhang M, Yang L, Zeng M. GPR68 Improves Nerve Damage and Myelination in an Immature Rat Model Induced by Sevoflurane Anesthesia by Activating cAMP/CREB to Mediate BDNF. ACS Chem Neurosci 2022; 13:423-431. [PMID: 35025202 DOI: 10.1021/acschemneuro.1c00830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer G-protein-coupled receptor 1 (OGR1, also known as GPR68) is a member of proton-sensing G-protein-coupled receptors, involved in cardiovascular physiology, tumor biology, and asthma, and exerts a neuroprotective effect against brain ischemia. The effects of GPR68 on anesthesia-induced nerve damage and myelination were investigated in this study. First, 2-day old postnatal rats were exposed to 4.9% sevoflurane for 2 h. Data from hematoxylin and eosin staining and Nissl staining showed that sevoflurane induced pathological changes in the hippocampus with a reduced number of neurons. GPR68 was downregulated in the hippocampus of sevoflurane-induced rats. Second, sevoflurane-induced rats were injected with adeno-associated virus (AAV)-mediated overexpression of GPR68, and overexpression of GPR68 ameliorated sevoflurane-induced pathological changes, enhanced the number of neurons, and improved the learning and memory function. Moreover, overexpression of GPR68 increased the number of BrdU-positive and Olig2-positive cells and enhanced protein expression of Olig2 in sevoflurane-induced rats. Third, the number of myelin basic protein (MBP) positive cells and protein expression of MBP in sevoflurane-induced rats were also enhanced by injection with AAV-GPR68. Overexpression of GPR68 attenuated sevoflurane-induced neuronal apoptosis and oxidative stress in rats. Lastly, overexpression of GPR68 upregulated protein expression of the brain-derived neurotrophic factor (BDNF) by increasing cAMP and phosphorylated cAMP response element-binding protein (CREB). In conclusion, GPR68 alleviated sevoflurane-induced nerve damage and myelination through BDNF-mediated activation of the cAMP/CREB pathway.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Anesthesiology, Chengdu University of Traditional Chinese Medicine Hospital, Chengdu, Sichuan 610032, China
| | - Minli Zhang
- Department of Anesthesiology, Chengdu University of Traditional Chinese Medicine Hospital, Chengdu, Sichuan 610032, China
| | - Lingling Yang
- Department of Anesthesiology, Chengdu University of Traditional Chinese Medicine Hospital, Chengdu, Sichuan 610032, China
| | - Mingquan Zeng
- Department of Critical Care Medicine, Public Health Clinical Center of Chengdu, Chengdu, Sichuan 610000, China
| |
Collapse
|
24
|
Liao Y, Wang J, Guo C, Bai M, Ju B, Ran Z, Hu J, Yang J, Wen A, Ding Y. Combination of Systems Pharmacology and Experimental Evaluation to Explore the Mechanism of Synergistic Action of Frankincense-Myrrh in the Treatment of Cerebrovascular Diseases. Front Pharmacol 2022; 12:796224. [PMID: 35082676 PMCID: PMC8784887 DOI: 10.3389/fphar.2021.796224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
Frankincense-Myrrh is a classic drug pair that promotes blood circulation, and eliminates blood stasis. The combination of the two drugs has a definite clinical effect on the treatment of cerebrovascular diseases (CBVDs), but its mechanism of action and compatibility have not been elucidated. In this study, the bioactive components, core targets, and possible synergistic mechanisms of Frankincense-Myrrh in the treatment of CBVDs are explored through systems pharmacology combined with in vivo and in vitro experiments. Comparing target genes of components in Frankincense and Myrrh with CBVD-related genes, common genes were identified; 15 core target genes of Frankincense-Myrrh for the treatment of CBVDs were then identified using protein-protein interaction (PPI) analysis. It was also predicted through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis that the molecular mechanism of Frankincense-Myrrh action on CBVDs was mainly related to the regulation of neurotrophic factors and inflammatory responses. Frankincense-Myrrh significantly improved neurological function, decreased infarct volume, alleviated histopathological damage, inhibited microglial expression, and promoted the expression of neurons in middle cerebral artery occlusion (MCAO)-induced rats. The results of this study not only provide important theoretical support and experimental basis for the synergistic effect of Frankincense-Myrrh, but also provide new ideas for the prevention and treatment of cerebral ischemic injuries.
Collapse
Affiliation(s)
- Yucheng Liao
- College of Pharmacy, Xinjiang Medical University, Urumqi, China.,Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Min Bai
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Bowei Ju
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Zheng Ran
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Junping Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Jianhua Yang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China.,Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
25
|
Yang L, Lei JF, Ouyang JY, Li MZ, Zhan Y, Feng XF, Lu Y, Li MC, Wang L, Zou HY, Zhao H. Effect of Neurorepair for Motor Functional Recovery Enhanced by Total Saponins From Trillium tschonoskii Maxim. Treatment in a Rat Model of Focal Ischemia. Front Pharmacol 2021; 12:763181. [PMID: 34955834 PMCID: PMC8703076 DOI: 10.3389/fphar.2021.763181] [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: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Trillium tschonoskii Maxim. (TTM), is a perennial herb from Liliaceae, that has been widely used as a traditional Chinese medicine treating cephalgia and traumatic hemorrhage. The present work was designed to investigate whether the total saponins from Trillium tschonoskii Maxim. (TSTT) would promote brain remodeling and improve gait impairment in the chronic phase of ischemic stroke. A focal ischemic model of male Sprague-Dawley (SD) rats was established by permanent middle cerebral artery occlusion (MCAO). Six hours later, rats were intragastrically treated with TSTT (120, 60, and 30 mg/kg) and once daily up to day 30. The gait changes were assessed by the CatWalk-automated gait analysis system. The brain tissues injuries, cerebral perfusion and changes of axonal microstructures were detected by multimodal magnetic resonance imaging (MRI), followed by histological examinations. The axonal regeneration related signaling pathways including phosphatidylinositol 3-kinases (PI3K)/protein kinase B (AKT)/glycogen synthase kinase-3 (GSK-3)/collapsin response mediator protein-2 (CRMP-2) were measured by western blotting. TSTT treatment significantly improved gait impairment of rats. MRI analysis revealed that TSTT alleviated tissues injuries, significantly improved cerebral blood flow (CBF), enhanced microstructural integrity of axon and myelin sheath in the ipsilesional sensorimotor cortex and internal capsule. In parallel to MRI findings, TSTT preserved myelinated axons and promoted oligodendrogenesis. Specifically, TSTT interventions markedly up-regulated expression of phosphorylated GSK-3, accompanied by increased expression of phosphorylated PI3K, AKT, but reduced phosphorylated CRMP-2 expression. Taken together, our results suggested that TSTT facilitated brain remodeling. This correlated with improving CBF, encouraging reorganization of axonal microstructure, promoting oligodendrogenesis and activating PI3K/AKT/GSK-3/CRMP-2 signaling, thereby improving poststroke gait impairments.
Collapse
Affiliation(s)
- Le Yang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Jian-Feng Lei
- Medical Imaging Laboratory of Core Facility Center, Capital Medical University, Beijing, China
| | - Jun-Yao Ouyang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Man-Zhong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yu Zhan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xue-Feng Feng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Ming-Cong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Hai-Yan Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| |
Collapse
|
26
|
Jurcau A, Simion A. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. Int J Mol Sci 2021; 23:14. [PMID: 35008440 PMCID: PMC8744548 DOI: 10.3390/ijms23010014] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Its increasing incidence has led stroke to be the second leading cause of death worldwide. Despite significant advances in recanalization strategies, patients are still at risk for ischemia/reperfusion injuries in this pathophysiology, in which neuroinflammation is significantly involved. Research has shown that in the acute phase, neuroinflammatory cascades lead to apoptosis, disruption of the blood-brain barrier, cerebral edema, and hemorrhagic transformation, while in later stages, these pathways support tissue repair and functional recovery. The present review discusses the various cell types and the mechanisms through which neuroinflammation contributes to parenchymal injury and tissue repair, as well as therapeutic attempts made in vitro, in animal experiments, and in clinical trials which target neuroinflammation, highlighting future therapeutic perspectives.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| | - Aurel Simion
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurorehabilitation Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| |
Collapse
|
27
|
Otero-Ortega L, Gutiérrez-Fernández M, Díez-Tejedor E. Recovery After Stroke: New Insight to Promote Brain Plasticity. Front Neurol 2021; 12:768958. [PMID: 34867756 PMCID: PMC8639681 DOI: 10.3389/fneur.2021.768958] [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: 09/01/2021] [Accepted: 10/18/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of Hospital La Paz Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of Hospital La Paz Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of Hospital La Paz Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
28
|
Alia C, Cangi D, Massa V, Salluzzo M, Vignozzi L, Caleo M, Spalletti C. Cell-to-Cell Interactions Mediating Functional Recovery after Stroke. Cells 2021; 10:3050. [PMID: 34831273 PMCID: PMC8623942 DOI: 10.3390/cells10113050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Ischemic damage in brain tissue triggers a cascade of molecular and structural plastic changes, thus influencing a wide range of cell-to-cell interactions. Understanding and manipulating this scenario of intercellular connections is the Holy Grail for post-stroke neurorehabilitation. Here, we discuss the main findings in the literature related to post-stroke alterations in cell-to-cell interactions, which may be either detrimental or supportive for functional recovery. We consider both neural and non-neural cells, starting from astrocytes and reactive astrogliosis and moving to the roles of the oligodendrocytes in the support of vulnerable neurons and sprouting inhibition. We discuss the controversial role of microglia in neural inflammation after injury and we conclude with the description of post-stroke alterations in pyramidal and GABAergic cells interactions. For all of these sections, we review not only the spontaneous evolution in cellular interactions after ischemic injury, but also the experimental strategies which have targeted these interactions and that are inspiring novel therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Claudia Alia
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| | - Daniele Cangi
- Department of Neurosciences, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, 50121 Florence, Italy;
| | - Verediana Massa
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| | - Marco Salluzzo
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
- Department of Neurosciences, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, 50121 Florence, Italy;
| | - Livia Vignozzi
- Department of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35121 Padua, Italy;
| | - Matteo Caleo
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
- Department of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35121 Padua, Italy;
| | - Cristina Spalletti
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| |
Collapse
|
29
|
Shaheryar ZA, Khan MA, Adnan CS, Zaidi AA, Hänggi D, Muhammad S. Neuroinflammatory Triangle Presenting Novel Pharmacological Targets for Ischemic Brain Injury. Front Immunol 2021; 12:748663. [PMID: 34691061 PMCID: PMC8529160 DOI: 10.3389/fimmu.2021.748663] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is one of the leading causes of morbidity and mortality globally. Hundreds of clinical trials have proven ineffective in bringing forth a definitive and effective treatment for ischemic stroke, except a myopic class of thrombolytic drugs. That, too, has little to do with treating long-term post-stroke disabilities. These studies proposed diverse options to treat stroke, ranging from neurotropic interpolation to venting antioxidant activity, from blocking specific receptors to obstructing functional capacity of ion channels, and more recently the utilization of neuroprotective substances. However, state of the art knowledge suggests that more pragmatic focus in finding effective therapeutic remedy for stroke might be targeting intricate intracellular signaling pathways of the 'neuroinflammatory triangle': ROS burst, inflammatory cytokines, and BBB disruption. Experimental evidence reviewed here supports the notion that allowing neuroprotective mechanisms to advance, while limiting neuroinflammatory cascades, will help confine post-stroke damage and disabilities.
Collapse
Affiliation(s)
- Zaib A. Shaheryar
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Faculty of Pharmacy, University of Lahore, Lahore, Pakistan
| | - Mahtab A. Khan
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan
| | | | - Awais Ali Zaidi
- Faculty of Pharmacy, University of Lahore, Lahore, Pakistan
- Imran Idrees College of Pharmacy, Lahore, Pakistan
| | - Daniel Hänggi
- Department of Neurosurgery, Faculty of Medicine and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Neurosurgery, Faculty of Medicine and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
30
|
Kim B, Schweighofer N, Haldar JP, Leahy RM, Winstein CJ. Corticospinal Tract Microstructure Predicts Distal Arm Motor Improvements in Chronic Stroke. J Neurol Phys Ther 2021; 45:273-281. [PMID: 34269747 PMCID: PMC8460613 DOI: 10.1097/npt.0000000000000363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE The corticospinal tract (CST) is a crucial brain pathway for distal arm and hand motor control. We aimed to determine whether a diffusion tensor imaging (DTI)-derived CST metric predicts distal upper extremity (UE) motor improvements in chronic stroke survivors. METHODS We analyzed clinical and neuroimaging data from a randomized controlled rehabilitation trial. Participants completed clinical assessments and neuroimaging at baseline and clinical assessments 4 months later, postintervention. Using univariate linear regression analysis, we determined the linear relationship between the DTI-derived CST fractional anisotropy asymmetry (FAasym) and the percentage of baseline change in log-transformed average Wolf Motor Function Test time for distal items (ΔlnWMFT-distal_%). The least absolute shrinkage and selection operator (LASSO) linear regressions with cross-validation and bootstrapping were used to determine the relative weighting of CST FAasym, other brain metrics, clinical outcomes, and demographics on distal motor improvement. Logistic regression analyses were performed to test whether the CST FAasym can predict clinically significant UE motor improvement. RESULTS lnWMFT-distal significantly improved at the group level. Baseline CST FAasym explained 26% of the variance in ΔlnWMFT-distal_%. A multivariate LASSO model including baseline CST FAasym, age, and UE Fugl-Meyer explained 39% of the variance in ΔlnWMFT-distal_%. Further, CST FAasym explained more variance in ΔlnWMFT-distal_% than the other significant predictors in the LASSO model. DISCUSSION AND CONCLUSIONS CST microstructure is a significant predictor of improvement in distal UE motor function in the context of an UE rehabilitation trial in chronic stroke survivors with mild-to-moderate motor impairment.Video Abstract available for more insight from the authors (see the Video, Supplemental Digital Content 1, available at: http://links.lww.com/JNPT/A350).
Collapse
Affiliation(s)
- Bokkyu Kim
- Department of Physical Therapy Education, SUNY Upstate Medical University, Syracuse, NY, United States
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, United States
| | - Nicolas Schweighofer
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, United States
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States
| | - Justin P. Haldar
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA, United States
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA, United States
| | - Richard M. Leahy
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA, United States
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA, United States
| | - Carolee J. Winstein
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, United States
- Department. of Neurology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
31
|
Schlecht A, Vallon M, Wagner N, Ergün S, Braunger BM. TGFβ-Neurotrophin Interactions in Heart, Retina, and Brain. Biomolecules 2021; 11:biom11091360. [PMID: 34572573 PMCID: PMC8464756 DOI: 10.3390/biom11091360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic insults to the heart and brain, i.e., myocardial and cerebral infarction, respectively, are amongst the leading causes of death worldwide. While there are therapeutic options to allow reperfusion of ischemic myocardial and brain tissue by reopening obstructed vessels, mitigating primary tissue damage, post-infarction inflammation and tissue remodeling can lead to secondary tissue damage. Similarly, ischemia in retinal tissue is the driving force in the progression of neovascular eye diseases such as diabetic retinopathy (DR) and age-related macular degeneration (AMD), which eventually lead to functional blindness, if left untreated. Intriguingly, the easily observable retinal blood vessels can be used as a window to the heart and brain to allow judgement of microvascular damages in diseases such as diabetes or hypertension. The complex neuronal and endocrine interactions between heart, retina and brain have also been appreciated in myocardial infarction, ischemic stroke, and retinal diseases. To describe the intimate relationship between the individual tissues, we use the terms heart-brain and brain-retina axis in this review and focus on the role of transforming growth factor β (TGFβ) and neurotrophins in regulation of these axes under physiologic and pathologic conditions. Moreover, we particularly discuss their roles in inflammation and repair following ischemic/neovascular insults. As there is evidence that TGFβ signaling has the potential to regulate expression of neurotrophins, it is tempting to speculate, and is discussed here, that cross-talk between TGFβ and neurotrophin signaling protects cells from harmful and/or damaging events in the heart, retina, and brain.
Collapse
|
32
|
Microglia as the Critical Regulators of Neuroprotection and Functional Recovery in Cerebral Ischemia. Cell Mol Neurobiol 2021; 42:2505-2525. [PMID: 34460037 DOI: 10.1007/s10571-021-01145-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.
Collapse
|
33
|
Youssef MI, Ma J, Chen Z, Hu WW. Potential therapeutic agents for ischemic white matter damage. Neurochem Int 2021; 149:105116. [PMID: 34229025 DOI: 10.1016/j.neuint.2021.105116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022]
Abstract
Ischemic white matter damage (WMD) is increasingly being considered as one of the major causes of neurological disorders in older adults and preterm infants. The functional consequences of WMD triggers a progressive cognitive decline and dementia particularly in patients with ischemic cerebrovascular diseases. Despite the major stride made in the pathogenesis mechanisms of ischemic WMD in the last century, effective medications are still not available. So, there is an urgent need to explore a promising approach to slow the progression or modify its pathological course. In this review, we discussed the animal models, the pathological mechanisms and the potential therapeutic agents for ischemic WMD. The development in the studies of anti-oxidants, free radical scavengers, anti-inflammatory or anti-apoptotic agents and neurotrophic factors in ischemic WMD were summarized. The agents which either alleviate oligodendrocyte damage or promote its proliferation or differentiation may have potential value for the treatment of ischemic WMD. Moreover, drugs with multifaceted protective activities or a wide therapeutic window may be optimal for clinical translation.
Collapse
Affiliation(s)
- Mahmoud I Youssef
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jing Ma
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Zhong Chen
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Wei-Wei Hu
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
34
|
Platelets as drivers of ischemia/reperfusion injury after stroke. Blood Adv 2021; 5:1576-1584. [PMID: 33687431 DOI: 10.1182/bloodadvances.2020002888] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide and, despite reperfusion either via thrombolysis or thrombectomy, stroke patients often suffer from lifelong disabilities. These persistent neurological deficits may be improved by treating the ischemia/reperfusion (I/R) injury that occurs following ischemic stroke. There are currently no approved therapies to treat I/R injury, and thus it is imperative to find new targets to decrease the burden of ischemic stroke and related diseases. Platelets, cell fragments from megakaryocytes, are primarily known for their role in hemostasis. More recently, investigators have studied the nonhemostatic role of platelets in inflammatory pathologies, such as I/R injury after ischemic stroke. In this review, we seek to provide an overview of how I/R can lead to platelet activation and how activated platelets, in turn, can exacerbate I/R injury after stroke. We will also discuss potential mechanisms by which platelets may ameliorate I/R injury.
Collapse
|
35
|
Guo YS, Yuan M, Han Y, Shen XY, Gao ZK, Bi X. Therapeutic Potential of Cytokines in Demyelinating Lesions After Stroke. J Mol Neurosci 2021; 71:2035-2052. [PMID: 33970426 DOI: 10.1007/s12031-021-01851-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022]
Abstract
White matter damage is a component of most human stroke and usually accounts for at least half of the lesion volume. Subcortical white matter stroke (WMS) accounts for 25% of all strokes and causes severe motor and cognitive dysfunction. The adult brain has a very limited ability to repair white matter damage. Pathological analysis shows that demyelination or myelin loss is the main feature of white matter injury and plays an important role in long-term sensorimotor and cognitive dysfunction. This suggests that demyelination is a major therapeutic target for ischemic stroke injury. An acute inflammatory reaction is triggered by brain ischemia, which is accompanied by cytokine production. The production of cytokines is an important factor affecting demyelination and myelin regeneration. Different cytokines have different effects on myelin damage and myelin regeneration. Exploring the role of cytokines in demyelination and remyelination after stroke and the underlying molecular mechanisms of demyelination and myelin regeneration after ischemic injury is very important for the development of rehabilitation treatment strategies. This review focuses on recent findings on the effects of cytokines on myelin damage and remyelination as well as the progress of research on the role of cytokines in ischemic stroke prognosis to provide a new treatment approach for amelioration of white matter damage after stroke.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Shanghai University of Sport, Shanghai, 200438, China
| | - Mei Yuan
- Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Han
- Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-Ya Shen
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Zhen-Kun Gao
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China.
| |
Collapse
|
36
|
Khan H, Pan JJ, Li Y, Zhang Z, Yang GY. Native and Bioengineered Exosomes for Ischemic Stroke Therapy. Front Cell Dev Biol 2021; 9:619565. [PMID: 33869170 PMCID: PMC8044840 DOI: 10.3389/fcell.2021.619565] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Exosomes are natural cells-derived vesicles, which are at the forefront toward clinical success for various diseases, including cerebral ischemia. Exosomes mediate cell-to-cell communication in different brain cells during both physiological and pathological conditions. Exosomes are an extensively studied type of extracellular vesicle, which are considered to be the best alternative for stem cell-based therapy. They can be secreted by various cell types and have unique biological properties. Even though native exosomes have potential for ischemic stroke therapy, some undesirable features prevent their success in clinical applications, including a short half-life, poor targeting property, low concentration at the target site, rapid clearance from the lesion region, and inefficient payload. In this review, we highlight exosome trafficking and cellular uptake and survey the latest discoveries in the context of exosome research as the best fit for brain targeting owing to its natural brain-homing abilities. Furthermore, we overview the methods by which researchers have bioengineered exosomes (BioEng-Exo) for stroke therapy. Finally, we summarize studies in which exosomes were bioengineered by a third party for stroke recovery. This review provides up-to-date knowledge about the versatile nature of exosomes with a special focus on BioEng-Exo for ischemic stroke. Standard exosome bioengineering techniques are mandatory for the future and will lead exosomes toward clinical success for stroke therapy.
Collapse
Affiliation(s)
- Haroon Khan
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jia-Ji Pan
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongfang Li
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
Cheng Q, Chen X, Ma J, Jiang X, Chen J, Zhang M, Wu Y, Zhang W, Chen C. Effect of Methylene Blue on White Matter Injury after Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6632411. [PMID: 33603949 PMCID: PMC7872771 DOI: 10.1155/2021/6632411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 12/04/2022]
Abstract
Methylene blue, the FDA-grandfathered drug was proved to be neuroprotective in ischemic stroke in rat. However, the mechanism of the protective effect was unknown. In this study, we used different animal models to investigate the effect of MB administration given within and beyond the therapeutic time window on behavioral deficits and infarct volume and related mechanism about the white matter protection. Middle cerebral artery occlusion and reperfusion (MCAO) and photothrombotic middle cerebral artery occlusion (PT-MCAO) models were used. Behavioral deficits and infarct volume were measured by foot fault test, Garcia neurological score, and TTC staining. Black gold staining and western blot were used to evaluate the brain white matter injury. We found that intraperitoneal administration of MB immediately or 24 h after the MCAO or PT-MCAO surgery reduced infarct volume, improved the neurological deficits, and reduced the white matter injury via myelin basic protein (BMP) protection. These findings suggested that MB relieved the white matter injury besides neuronal protection and has potential therapeutic effects on ischemic stroke.
Collapse
Affiliation(s)
- Quancheng Cheng
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Xuhao Chen
- School of Clinical Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Jiayi Ma
- School of Clinical Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Xingyuan Jiang
- School of Clinical Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Jiahui Chen
- School of Clinical Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Mengqin Zhang
- School of Clinical Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Yejun Wu
- School of Clinical Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Weiguang Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing 100191, China
| |
Collapse
|
38
|
Gou X, Ying J, Yue Y, Qiu X, Hu P, Qu Y, Li J, Mu D. The Roles of High Mobility Group Box 1 in Cerebral Ischemic Injury. Front Cell Neurosci 2020; 14:600280. [PMID: 33384585 PMCID: PMC7770223 DOI: 10.3389/fncel.2020.600280] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that plays an important role in stabilizing nucleosomes and DNA repair. HMGB1 can be passively released from necrotic neurons or actively secreted by microglia, macrophages/monocytes, and neutrophils. Cerebral ischemia is a major cause of mortality and disability worldwide, and its outcome depends on the number of neurons dying due to hypoxia in the ischemic area. HMGB1 contributes to the pathogenesis of cerebral ischemia via mediating neuroinflammatory responses to cerebral ischemic injury. Extracellular HMGB1 regulates many neuroinflammatory events by interacting with its different cell surface receptors, such as receptors for advanced glycation end products, toll-like receptor (TLR)-2, and TLR-4. Additionally, HMGB1 can be redox-modified, thus exerting specific cellular functions in the ischemic brain and has different roles in the acute and late stages of cerebral ischemic injury. However, the role of HMGB1 in cerebral ischemia is complex and remains unclear. Herein, we summarize and review the research on HMGB1 in cerebral ischemia, focusing especially on the role of HMGB1 in hypoxic ischemia in the immature brain and in white matter ischemic injury. We also outline the possible mechanisms of HMGB1 in cerebral ischemia and the main strategies to inhibit HMGB1 pertaining to its potential as a novel critical molecular target in cerebral ischemic injury.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Yue
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Peng Hu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Free Fatty Acids and Their Inflammatory Derivatives Affect BDNF in Stroke Patients. Mediators Inflamm 2020; 2020:6676247. [PMID: 33343231 PMCID: PMC7728491 DOI: 10.1155/2020/6676247] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/18/2022] Open
Abstract
Objective The neurotrophin brain-derived neurotrophic factor (BDNF) affects poststroke functional outcome, neurogenesis, neuroprotection, and neuroplasticity. Its level is related to the diet and nutritional status, and more specifically, it is free fatty acids (FFAs) and eicosanoids that can have an impact on the BDNF level. The aim of this study was to analyze the potential impact of FFAs and eicosanoids on the BDNF level in stroke patients. Material and Methods. Seventy-three ischemic stroke patients were prospectively enrolled in the study. Laboratory tests were performed in all subjects, including the levels of FFAs, eicosanoids, and BDNF. FFAs and inflammatory metabolites were determined by gas chromatography and liquid chromatography, while BDNF was evaluated by the immune-enzymatic method (ELISA). Results The plasma level of BDNF negatively correlated with C22:1n9 13 erucic acid, C18:3n3 linolenic acid (ALA), and lipoxin A4 15-epi-LxA4. A direct association was observed in relation to BDNF and C16:1 palmitoleic acid and C20:3n6 eicosatrienoic acid (dihomo-gamma-linolenic acid (DGLA)). Conclusions Saturated fatty acids and omega-3 and omega-9 erucic acids can affect signaling in the BDNF synthesis resulting in the decrease in BDNF. There is a beneficial effect of DGLA on the BDNF level, while the effect of ALA on BDNF can be inhibitory. Specialized proresolving lipid mediators can play a role in the BDNF metabolism. BDNF can interact with inflammation as the risk factor in the cardiovascular disorders, including stroke.
Collapse
|
40
|
Qiu H, Qian T, Wu T, Wang X, Zhu C, Chen C, Wang L. Umbilical cord blood cells for the treatment of preterm white matter injury: Potential effects and treatment options. J Neurosci Res 2020; 99:778-792. [PMID: 33207392 DOI: 10.1002/jnr.24751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022]
Abstract
Preterm birth is a global public health problem. A large number of preterm infants survive with preterm white matter injury (PWMI), which leads to neurological deficits, and has multifaceted etiology, clinical course, monitoring, and outcomes. The principal upstream insults leading to PWMI initiation are hypoxia-ischemia and infection and/or inflammation and the key target cells are late oligodendrocyte precursor cells. Current PWMI treatments are mainly supportive, and thus have little effect in terms of protecting the immature brain or repairing injury to improve long-term outcomes. Umbilical cord blood (UCB) cells comprise abundant immunomodulatory and stem cells, which have the potential to reduce brain injury, mainly due to anti-inflammatory and immunomodulatory mechanisms, and also through their release of neurotrophic or growth factors to promote endogenous neurogenesis. In this review, we briefly summarize PWMI pathogenesis and pathophysiology, and the specific properties of different cell types in UCB. We further explore the potential mechanism by which UCB can be used to treat PWMI, and discuss the advantages of and potential issues related to UCB cell therapy. Finally, we suggest potential future studies of UCB cell therapy in preterm infants.
Collapse
Affiliation(s)
- Han Qiu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tianyang Qian
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tong Wu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoyang Wang
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Chao Chen
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Laishuan Wang
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
41
|
Colucci-D’Amato L, Speranza L, Volpicelli F. Neurotrophic Factor BDNF, Physiological Functions and Therapeutic Potential in Depression, Neurodegeneration and Brain Cancer. Int J Mol Sci 2020; 21:E7777. [PMID: 33096634 PMCID: PMC7589016 DOI: 10.3390/ijms21207777] [Citation(s) in RCA: 424] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 01/10/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most distributed and extensively studied neurotrophins in the mammalian brain. BDNF signals through the tropomycin receptor kinase B (TrkB) and the low affinity p75 neurotrophin receptor (p75NTR). BDNF plays an important role in proper growth, development, and plasticity of glutamatergic and GABAergic synapses and through modulation of neuronal differentiation, it influences serotonergic and dopaminergic neurotransmission. BDNF acts as paracrine and autocrine factor, on both pre-synaptic and post-synaptic target sites. It is crucial in the transformation of synaptic activity into long-term synaptic memories. BDNF is considered an instructive mediator of functional and structural plasticity in the central nervous system (CNS), influencing dendritic spines and, at least in the hippocampus, the adult neurogenesis. Changes in the rate of adult neurogenesis and in spine density can influence several forms of learning and memory and can contribute to depression-like behaviors. The possible roles of BDNF in neuronal plasticity highlighted in this review focus on the effect of antidepressant therapies on BDNF-mediated plasticity. Moreover, we will review data that illustrate the role of BDNF as a potent protective factor that is able to confer protection against neurodegeneration, in particular in Alzheimer's disease. Finally, we will give evidence of how the involvement of BDNF in the pathogenesis of brain glioblastoma has emerged, thus opening new avenues for the treatment of this deadly cancer.
Collapse
Affiliation(s)
- Luca Colucci-D’Amato
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
- InterUniversity Center for Research in Neurosciences (CIRN), University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Luisa Speranza
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| |
Collapse
|
42
|
Kalinichenko SG, Matveeva NY, Korobtsov AV. Brain-Derived Neurotrophic Factor (BDNF) As a Regulator of Apoptosis under Conditions of Focal Experimental Stroke. Bull Exp Biol Med 2020; 169:701-706. [PMID: 32990850 DOI: 10.1007/s10517-020-04959-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Indexed: 11/25/2022]
Abstract
The immunolocalization of apoptotic factors in rat neocortex was studied on the model of permanent occlusion of the middle cerebral artery with administration of exogenous BDNF. We revealed heterogeneous distribution of pro- and anti-apoptotic factors in the stroke area and in the surrounding penumbra, where caspase-3+ and p53+ cells were found. Their number was maximum on day 3 of ischemia. The number of neurons containing anti-apoptotic factors was relatively decreased. Injection of BDNF changed the distribution of apoptotic factors. In the penumbra area, BDNF enhanced the expression of Mdm2 primarily in the pyramid cells of layers V/VI and Bcl-2 in interneurons of layers II and III. Localization of p53 and caspase-3 varied at different terms of the ischemic period and showed an inverse dependence. Considering the selective neuroprotective effect of BDNF, various mechanisms of the formation of ischemic tolerance in neurons are proposed.
Collapse
Affiliation(s)
- S G Kalinichenko
- Department of Histology, Cytology and Embryology, Pacific State Medical University, Vladivostok, Russia.
| | - N Y Matveeva
- Department of Histology, Cytology and Embryology, Pacific State Medical University, Vladivostok, Russia
| | - A V Korobtsov
- Department of Histology, Cytology and Embryology, Pacific State Medical University, Vladivostok, Russia
| |
Collapse
|
43
|
He J, Huang Y, Liu H, Sun X, Wu J, Zhang Z, Liu L, Zhou C, Jiang S, Huang Z, Zhong J, Guo Z, Jiang L, Cheng C. Bexarotene promotes microglia/macrophages - Specific brain - Derived Neurotrophic factor expression and axon sprouting after traumatic brain injury. Exp Neurol 2020; 334:113462. [PMID: 32916173 DOI: 10.1016/j.expneurol.2020.113462] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 08/14/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022]
Abstract
Traumatic brain injury (TBI) has been regarded as one of the leading cause of injury-related death and disability. White matter injury after TBI is characterized by axon damage and demyelination, resulting in neural network impairment and neurological deficit. Brain-derived neurotrophic factor (BDNF) can promote white matter repair. The activation of peroxisome proliferator-activated receptor gamma (PPARγ) has been reported to promote microglia/macrophages towards anti-inflammatory state and therefore to promote axon regeneration. Bexarotene, an agonist of retinoid X receptor (RXR), can activate RXR/PPARγ heterodimers. The aim of the present study was to identify the effect of bexarotene on BDNF in microglia/macrophages and axon sprouting after TBI in mice. Bexarotene was administered intraperitoneally in C57BL/6 mice undergoing controlled cortical impact (CCI). PPARγ dependency was determined by intraperitoneal administration of a PPARγ antagonist T0070907. We found that bexarotene promoted axon regeneration indicated by increased growth associated protein 43 (GAP43) expression, myelin basic protein (MBP) expression, and biotinylated dextran amine (BDA)+ axon sprouting. Bexarotene also increased microglia/macrophages-specific brain derived neurotrophic factor (BDNF) expression after TBI. In addition, bexarotene reduced the number of pro-inflammatory microglia/macrophages while increased the number of anti-inflammatory microglia/macrophages after TBI. Moreover, bexaortene inhibited pro-inflammatory cytokine secretion. In addition, bexarotene treatment improved neurological scores and cognitive function of CCI-injured mice. These effects of bexarotene were partially abolished by T0070907. In conclusion, bexarotene promotes axon sprouting, increases microglia/macrophages-specific BDNF expression, and induces microglia/macrophages from a pro-inflammatory state towards an anti-inflammatory one after TBI at least partially in a PPARγ-dependent manner.
Collapse
Affiliation(s)
- Junchi He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yike Huang
- Department of Ophthalmology, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, China
| | - Han Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingchuan Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaosi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shaoqiu Jiang
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhijian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zongduo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
44
|
Li L, Li R, Zacharek A, Wang F, Landschoot-Ward J, Chopp M, Chen J, Cui X. ABCA1/ApoE/HDL Signaling Pathway Facilitates Myelination and Oligodendrogenesis after Stroke. Int J Mol Sci 2020; 21:ijms21124369. [PMID: 32575457 PMCID: PMC7352241 DOI: 10.3390/ijms21124369] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) plays an important role in the regulation of apolipoprotein E (ApoE) and the biogenesis of high-density lipoprotein (HDL) cholesterol in the mammalian brain. Cholesterol is a major source for myelination. Here, we investigate whether ABCA1/ApoE/HDL contribute to myelin repair and oligodendrogenesis in the ischemic brain after stroke. Specific brain ABCA1-deficient (ABCA1-B/-B) and ABCA1-floxed (ABCA1fl/fl) control mice were subjected to permanent distal middle-cerebral-artery occlusion (dMCAo) and were intracerebrally administered (1) artificial mouse cerebrospinal fluid (CSF) as vehicle control, (2) human plasma HDL3, and (3) recombined human ApoE2 starting 24 h after dMCAo for 14 days. All stroke mice were sacrificed 21 days after dMCAo. The ABCA1-B/-B–dMCAo mice exhibit significantly reduced myelination and oligodendrogenesis in the ischemic brain as well as decreased functional outcome 21 days after stroke compared with ABCA1fl/fl mice; administration of human ApoE2 or HDL3 in the ischemic brain significantly attenuates the deficits in myelination and oligodendrogenesis in ABCA1-B/-B–dMCAo mice ( p < 0.05, n = 9/group). In vitro, ABCA1-B/-B reduces ApoE expression and decreases primary oligodendrocyte progenitor cell (OPC) migration and oligodendrocyte maturation; HDL3 and ApoE2 treatment significantly reverses ABCA1-B/-B-induced reduction in OPC migration and oligodendrocyte maturation. Our data indicate that the ABCA1/ApoE/HDL signaling pathway contributes to myelination and oligodendrogenesis in the ischemic brain after stroke.
Collapse
Affiliation(s)
- Li Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Rongwen Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Fengjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Julie Landschoot-Ward
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Xu Cui
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
- Correspondence: ; Tel.: 01-313-916-2864
| |
Collapse
|
45
|
Epimedium flavonoids improve cognitive impairment and white matter lesions induced by chronic cerebral hypoperfusion through inhibiting the Lingo-1/Fyn/ROCK pathway and activating the BDNF/NRG1/PI3K pathway in rats. Brain Res 2020; 1743:146902. [PMID: 32446949 DOI: 10.1016/j.brainres.2020.146902] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/09/2020] [Accepted: 05/18/2020] [Indexed: 01/24/2023]
Abstract
Chronic cerebral hypoperfusion is a common cause of cerebral small vascular disease (CSVD). White matter (WM) lesions are the typical pathological manifestation of CSVD and contribute to cognitive decline. Epimedium flavonoids (EF) are the main component in Epimedium brevicornu Maxim., which is commonly used in traditional Chinese medicine. The purpose of this study was to investigate the effects of EF on cognitive impairment and the underlying mechanisms in a CSVD rat model induced with chronic cerebral hypoperfusion. The model was established by permanent bilateral common carotid artery occlusion (2VO) in rats. EF (50, 100, and 200 mg/kg) was intragastrically administered once a day for 12 weeks starting 2 weeks after 2VO surgery. The learning and memory capacity of the rats were measured using the Morris water maze and step-through tests. WM lesions were observed by MRI-diffusion tensor imaging, transmission electron microscopy, and LFB staining. Oligodendrocytes were detected by immunohistochemistry. Western blotting assay was used to determine the level of protein expression. The results showed that EF significantly improved learning and memory impairment, alleviated WM nerve fiber injuries and demyelination, and increased the number of mature oligodendrocytes in the corpus callosum, subcortical WM, and periventricular WM in 2VO rats. Mechanistically, EF reduced the expression of Lingo-1 and ROCK2 and increased the levels of phosphorylated (p-) Fyn, brain-derived neurotrophic factor (BDNF), TrkB, neuregulin-1 (NRG-1), p-ErbB4, PI3K p85 and p110α, p-Akt, and p-CREB in the corpus callosum of 2VO rats. These results suggest that EF may improve cognitive impairment and WM lesions induced by chronic cerebral hypoperfusion through inhibiting the Lingo-1/Fyn/ROCK pathway and activating the BDNF/TrkB, NRG-1/ErbB4, and the downstream PI3K/Akt/CREB pathways in WM. Thus, EF can be used as a potential neuroprotective agent in CSVD therapy.
Collapse
|
46
|
Li M, Xia M, Chen W, Wang J, Yin Y, Guo C, Li C, Tang X, Zhao H, Tan Q, Chen Y, Jia Z, Liu X, Feng H. Lithium treatment mitigates white matter injury after intracerebral hemorrhage through brain-derived neurotrophic factor signaling in mice. Transl Res 2020; 217:61-74. [PMID: 31951826 DOI: 10.1016/j.trsl.2019.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/04/2023]
Abstract
Intracerebral hemorrhage (ICH), a subtype of stroke with high morbidity and mortality, occurs mainly in the basal ganglia and causes white matter injury (WMI), resulting in severe motor dysfunction and poor prognosis in patients. The preservation of the white matter around the hematoma is crucial for motor function recovery, but there is currently no effective treatment for WMI following ICH. Lithium has been widely used for the treatment of bipolar disorder for decades. Although the protective effects of lithium on neurodegenerative diseases and cerebral trauma have been studied in recent years, whether it can be used to alleviate WMI after ICH remains to be researched. The results of this study revealed that ICH caused significant functional and pathological abnormalities in mice. After LiCl was administered to mice with ICH, behavioural performance and electrophysiological functions were improved and ICH-induced white matter pathological injury, including myelin sheath and axonal degeneration, was ameliorated. Furthermore, LiCl treatment decreased the death of mature oligodendrocytes (OLGs) in ICH mice, which may have been attributed to the enhanced expression of brain-derived neurotrophic factor (BDNF) regulated by the LiCl-induced inhibition of glycogen synthase kinase-3β (GSK-3β). The decreased death of OLGs was closely associated with decreased destruction of the myelin sheath and alleviated degradation of the axons. In summary, this study suggests that the protective effect of lithium on WMI after ICH might be related to an increased level of BDNF and that LiCl treatment may be a potential therapeutic method to palliate WMI after ICH.
Collapse
Affiliation(s)
- Mingxi Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Min Xia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Jie Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yi Yin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chengcheng Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xiaoqin Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Hengli Zhao
- Department of Neurology, The Second Medical Central, Chinese PLA (People's Liberation Army) General Hospital, Beijing, PR China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; State Key Laboratory of Trauma, Burn, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Zhengcai Jia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China.
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; State Key Laboratory of Trauma, Burn, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, PR China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China.
| |
Collapse
|
47
|
Ronzano R, Thetiot M, Lubetzki C, Desmazieres A. Myelin Plasticity and Repair: Neuro-Glial Choir Sets the Tuning. Front Cell Neurosci 2020; 14:42. [PMID: 32180708 PMCID: PMC7059744 DOI: 10.3389/fncel.2020.00042] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/12/2020] [Indexed: 12/11/2022] Open
Abstract
The plasticity of the central nervous system (CNS) in response to neuronal activity has been suggested as early as 1894 by Cajal (1894). CNS plasticity has first been studied with a focus on neuronal structures. However, in the last decade, myelin plasticity has been unraveled as an adaptive mechanism of importance, in addition to the previously described processes of myelin repair. Indeed, it is now clear that myelin remodeling occurs along with life and adapts to the activity of neuronal networks. Until now, it has been considered as a two-part dialog between the neuron and the oligodendroglial lineage. However, other glial cell types might be at play in myelin plasticity. In the present review, we first summarize the key structural parameters for myelination, we then describe how neuronal activity modulates myelination and finally discuss how other glial cells could participate in myelinic adaptivity.
Collapse
Affiliation(s)
- Remi Ronzano
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
| | - Melina Thetiot
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
- Unit Zebrafish Neurogenetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, CNRS, Paris, France
| | - Catherine Lubetzki
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Anne Desmazieres
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
| |
Collapse
|
48
|
Chen FT, Erickson KI, Huang H, Chang YK. The association between physical fitness parameters and white matter microstructure in older adults: A diffusion tensor imaging study. Psychophysiology 2020; 57:e13539. [PMID: 32030777 DOI: 10.1111/psyp.13539] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/29/2019] [Accepted: 01/09/2020] [Indexed: 12/18/2022]
Abstract
The present study was designed to examine whether different measures of physical fitness are differentially associated with white matter (WM) microstructure in older adults. Fifty-six healthy adults (mean age: 59.14 years) completed a standardized evaluation of physical fitness measurements (e.g., VO2peak , push-ups, abdominal sit-ups, sit-and-reach, t test, and vertical jump). Fractional anisotropy (FA), an index of WM microstructure, was assessed using diffusion tensor imaging. The findings indicated that the cardiorespiratory fitness was positively associated with FA in the right cingulum hippocampus and the left cerebral peduncle. However, other physical fitness metrics were not significantly associated with FA in any region. These results suggest that cardiorespiratory fitness, but not other metrics of fitness, might be sensitive to WM microstructure.
Collapse
Affiliation(s)
- Feng-Tzu Chen
- Graduate Institute of Sport, Leisure and Hospitality Management, National Taiwan Normal University, Taipei, Republic of China (Taiwan)
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA.,Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Australia
| | - Haiqing Huang
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yu-Kai Chang
- Department of Physical Education, National Taiwan Normal University, Taipei, Republic of China (Taiwan).,Institute for Research Excellence in Learning Science, National Taiwan Normal University, Taipei, Republic of China (Taiwan)
| |
Collapse
|
49
|
Chen SY, Lin MC, Tsai JS, He PL, Luo WT, Chiu IM, Herschman HR, Li HJ. Exosomal 2',3'-CNP from mesenchymal stem cells promotes hippocampus CA1 neurogenesis/neuritogenesis and contributes to rescue of cognition/learning deficiencies of damaged brain. Stem Cells Transl Med 2020; 9:499-517. [PMID: 31943851 PMCID: PMC7103625 DOI: 10.1002/sctm.19-0174] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/25/2019] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in clinical studies to treat neurological diseases and damage. However, implanted MSCs do not achieve their regenerative effects by differentiating into and replacing neural cells. Instead, MSC secretome components mediate the regenerative effects of MSCs. MSC-derived extracellular vesicles (EVs)/exosomes carry cargo responsible for rescuing brain damage. We previously showed that EP4 antagonist-induced MSC EVs/exosomes have enhanced regenerative potential to rescue hippocampal damage, compared with EVs/exosomes from untreated MSCs. Here we show that EP4 antagonist-induced MSC EVs/exosomes promote neurosphere formation in vitro and increase neurogenesis and neuritogenesis in damaged hippocampi; basal MSC EVs/exosomes do not contribute to these regenerative effects. 2',3'-Cyclic nucleotide 3'-phosphodiesterase (CNP) levels in EP4 antagonist-induced MSC EVs/exosomes are 20-fold higher than CNP levels in basal MSC EVs/exosomes. Decreasing elevated exosomal CNP levels in EP4 antagonist-induced MSC EVs/exosomes reduced the efficacy of these EVs/exosomes in promoting β3-tubulin polymerization and in converting toxic 2',3'-cAMP into neuroprotective adenosine. CNP-depleted EP4 antagonist-induced MSC EVs/exosomes lost the ability to promote neurogenesis and neuritogenesis in damaged hippocampi. Systemic administration of EV/exosomes from EP4 -antagonist derived MSC EVs/exosomes repaired cognition, learning, and memory deficiencies in mice caused by hippocampal damage. In contrast, CNP-depleted EP4 antagonist-induced MSC EVs/exosomes failed to repair this damage. Exosomal CNP contributes to the ability of EP4 antagonist-elicited MSC EVs/exosomes to promote neurogenesis and neuritogenesis in damaged hippocampi and recovery of cognition, memory, and learning. This experimental approach should be generally applicable to identifying the role of EV/exosomal components in eliciting a variety of biological responses.
Collapse
Affiliation(s)
- Shih-Yin Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Meng-Chieh Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Jia-Shiuan Tsai
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Lin He
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Ting Luo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Harvey R Herschman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California.,Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, California.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Hua-Jung Li
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
50
|
Blocking the Thrombin Receptor Promotes Repair of Demyelinated Lesions in the Adult Brain. J Neurosci 2020; 40:1483-1500. [PMID: 31911460 DOI: 10.1523/jneurosci.2029-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 01/14/2023] Open
Abstract
Myelin loss limits neurological recovery and myelin regeneration and is critical for restoration of function. We recently discovered that global knock-out of the thrombin receptor, also known as Protease Activated Receptor 1 (PAR1), accelerates myelin development. Here we demonstrate that knocking out PAR1 also promotes myelin regeneration. Outcomes in two unique models of myelin injury and repair, that is lysolecithin or cuprizone-mediated demyelination, showed that PAR1 knock-out in male mice improves replenishment of myelinating cells and remyelinated nerve fibers and slows early axon damage. Improvements in myelin regeneration in PAR1 knock-out mice occurred in tandem with a skewing of reactive astrocyte signatures toward a prorepair phenotype. In cell culture, the promyelinating effects of PAR1 loss of function are consistent with possible direct effects on the myelinating potential of oligodendrocyte progenitor cells (OPCs), in addition to OPC-indirect effects involving enhanced astrocyte expression of promyelinating factors, such as BDNF. These findings highlight previously unrecognized roles of PAR1 in myelin regeneration, including integrated actions across the oligodendrocyte and astroglial compartments that are at least partially mechanistically linked to the powerful BDNF-TrkB neurotrophic signaling system. Altogether, findings suggest PAR1 may be a therapeutically tractable target for demyelinating disorders of the CNS.SIGNIFICANCE STATEMENT Replacement of oligodendroglia and myelin regeneration holds tremendous potential to improve function across neurological conditions. Here we demonstrate Protease Activated Receptor 1 (PAR1) is an important regulator of the capacity for myelin regeneration across two experimental murine models of myelin injury. PAR1 is a G-protein-coupled receptor densely expressed in the CNS, however there is limited information regarding its physiological roles in health and disease. Using a combination of PAR1 knock-out mice, oligodendrocyte monocultures and oligodendrocyte-astrocyte cocultures, we demonstrate blocking PAR1 improves myelin production by a mechanism related to effects across glial compartments and linked in part to regulatory actions toward growth factors such as BDNF. These findings set the stage for development of new clinically relevant myelin regeneration strategies.
Collapse
|