1
|
Chen M, Wang H, Chen P, Zhu G, Li S, Li Z, Liu X, Ye G, Chen W. Neonatal microglia transplantation at early stage but not late stage after traumatic brain injury shows protective effects in mice. J Neurophysiol 2024; 131:598-606. [PMID: 38380844 DOI: 10.1152/jn.00006.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 02/22/2024] Open
Abstract
The transplantation of neonatal microglia suppresses neuroinflammation caused by traumatic brain injury (TBI). This research aimed to explore the optimal time point of neonatal microglia transplantation for the best effect on the improvement of long-term cognitive function and inflammatory response in mouse models. qPCR and immunoblotting showed that the level of Iba1 gradually increased to the highest on day 7 and then gradually declined in TBI mice. Furthermore, it was observed that the level of CD86 and TNF-α increased to the highest after 7 days and subsequently was maintained until day 21, whereas the level of CD206 and IL-10 increased to the highest after 24 h and subsequently decreased until day 21 by qPCR and enzyme-linked immunosorbent assay. Afterward, it was shown that the neonatal microglia transplantation within 1 h significantly attenuated anxiety-like behavior and improved cognitive impairments in TBI mice. Mechanism exploration showed that the neonatal microglia could significantly decrease the level of cleaved caspase-3, M1/M2 polarization, and inflammatory cytokine (TNF-α) while increasing the level of anti-inflammatory factor IL-10 in TBI mice after transplantation within 1 h. Here, our findings demonstrated that neonatal microglia transplantation within 1 h significantly attenuated anxiety-like behavior and cognitive impairments caused by TBI.NEW & NOTEWORTHY The study demonstrated that neonatal microglia transplantation within 1 h significantly inhibited the pathogenesis of traumatic brain injury (TBI) in mouse models through inhibition of M1 polarization and promotion of M2 polarization.
Collapse
Affiliation(s)
- Maosong Chen
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hongcai Wang
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Pandi Chen
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guangyao Zhu
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shiwei Li
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zengpan Li
- Department of Emergency and Trauma Center, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xuelan Liu
- Department of Emergency and Trauma Center, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Gengfan Ye
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wei Chen
- Department of Neurosurgery, the Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Pudong New Area, Shanghai, China
| |
Collapse
|
2
|
Fatemeh B, Koorosh S, Amir S, Yaghoub F, Javad MZ. Intra-hippocampal cis-P tau microinjection induces long-term changes in behavior and synaptic plasticity in mice. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:9. [PMID: 37231523 DOI: 10.1186/s12993-023-00211-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Alzheimer's disease is accompanied by an abnormal high accumulation of cis-P tau. However, the long-term changes in behavior following tau accumulation remains under debate. The present study investigated the long-term effects of tauopathy on learning and memory, synaptic plasticity, and hippocampal cell numbers. RESULTS Cis-P tau was microinjected into the dorsal hippocampus to generate Alzheimer's like-disease model in C57BL/6 mice. Cis-P tau injected animals showed a significant impairment in learning and memory in Y-maze and Barnes maze tests. In another group of animals, the generation of long-term potentiation (LTP) was evaluated in hippocampal slices 7 months after cis-P tau injection. LTP induction was disrupted only in the dorsal but not ventral hippocampal slices. The basal synaptic transmission was also reduced in dorsal hippocampal slices. In addition, hippocampal sampling was done, and the number of cells was assessed by Nissl staining. Obtained results indicated that the number of survived cells was significantly reduced in the dorsal and ventral hippocampus of cis P-tau injected animals compared to the animals in control group. However, the decrement of cell number was higher in the dorsal compared to the ventral hippocampus. CONCLUSIONS In conclusion, intra-hippocampal cis-P tau injection produced learning and memory impairment at 7 months after its injection. This impairment might result from LTP disruption and a significant decrease in the number of neurons in the dorsal hippocampus.
Collapse
Affiliation(s)
- Bakhtiarzadeh Fatemeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran
| | - Shahpasand Koorosh
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shojaei Amir
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Fathollahi Yaghoub
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran
| | - Mirnajafi-Zadeh Javad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran.
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Wang Y, Chen Q, Dang X, Lu W, Zhang X, Yan H, Niu S, Yan X, Yan J. A bibliometric analysis on traumatic brain injury in forensic medicine of a half-century (1972-2021). Front Neurol 2023; 14:913855. [PMID: 36816552 PMCID: PMC9932540 DOI: 10.3389/fneur.2023.913855] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Traumatic brain injury (TBI) is among the most common injuries in forensic medicine, the identification of which is of particular importance in forensic practice. To reveal the circumstances and trends of TBI in the forensic field, we used the Web of Science (WoS) database for comprehensive retrieval. We made a metrological analysis of 1,089 papers in the past 50 years (1972-2021). The United States and Germany have the most forensic research on TBI. Diffuse axonal injury (DAI) has been the focus of attention for many years, and much effort has been devoted to its diagnosis in forensic pathology. Infants and children are the subgroups of most concern, especially in infant and child abuse cases. Research on identifying shaken baby syndrome has received increasing attention in recent years. Overall, our study provides a comprehensive list and analysis of the articles regarding TBI in legal medicine, which may shed light on recognizing the trends and research hotspots in this field.
Collapse
Affiliation(s)
- Yufang Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qianqian Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xingxing Dang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Wanqing Lu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xinran Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - He Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shuliang Niu
- School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Xisheng Yan
- Department of Cardiovascular Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China,School of Basic Medical Science, Xinjiang Medical University, Urumqi, China,*Correspondence: Jie Yan ✉
| |
Collapse
|
4
|
Katsumoto A, Kokiko-Cochran ON, Bemiller SM, Xu G, Ransohoff RM, Lamb BT. Triggering receptor expressed on myeloid cells 2 deficiency exacerbates injury-induced inflammation in a mouse model of tauopathy. Front Immunol 2022; 13:978423. [PMID: 36389767 PMCID: PMC9664165 DOI: 10.3389/fimmu.2022.978423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/14/2022] [Indexed: 01/24/2023] Open
Abstract
Traumatic brain injury (TBI) promotes several Alzheimer's disease-like pathological features, including microtubule-associated protein tau (MAPT) accumulation within neurons. Macrophage activation in the injured hTau mouse model of tauopathy raises the question whether there is a relationship between MAPT pathology and alterations in macrophage activation following TBI. Triggering receptor expressed on myeloid cells 2 (TREM2) is a critical regulator of microglia and macrophage phenotype, but its mechanisms on TBI remain unclear. To address the association with TREM2 in TBI and MAPT pathology, we studied TREM2 deficiency in hTau mice (hTau;Trem2-/- ) 3 (acute phase) and 120 (chronic phase) days after experimental TBI. At three days following injury, hTau;Trem2-/- mice exhibited reduced macrophage activation both in the cortex and hippocampus. However, to our surprise, hTau;Trem2-/- mice exposed to TBI augments macrophage accumulation in the corpus callosum and white matter near the site of tissue damage in a chronic phase, which results in exacerbated axonal injury, tau aggregation, and impaired neurogenesis. We further demonstrate that TREM2 deficiency in hTau injured mice promotes neuronal dystrophy in the white matter due to impaired phagocytosis of apoptotic cells. Remarkably, hTau;Trem2-/- exposed to TBI failed to restore blood-brain barrier integrity. These findings imply that TREM2 deficiency accelerates inflammation and neurodegeneration, accompanied by attenuated microglial phagocytosis and continuous blood-brain barrier (BBB) leakage, thus exacerbating tauopathy in hTau TBI mice.
Collapse
Affiliation(s)
- Atsuko Katsumoto
- Department of Neurosciences, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Olga N. Kokiko-Cochran
- Department of Neurosciences, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States,Department of Neurosciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shane M. Bemiller
- Department of Neurosciences, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Guixiang Xu
- Department of Neurosciences, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Richard M. Ransohoff
- Department of Neurosciences, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States,Neuroinflammation Research Center, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Bruce T. Lamb
- Department of Neurosciences, The Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States,*Correspondence: Bruce T. Lamb,
| |
Collapse
|
5
|
Guillemain G, Lacapere JJ, Khemtemourian L. Targeting hIAPP fibrillation: A new paradigm to prevent β-cell death? BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184002. [PMID: 35868406 DOI: 10.1016/j.bbamem.2022.184002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
Loss of pancreatic β-cell mass is deleterious for type 2 diabetes patients since it reduces insulin production, critical for glucose homeostasis. The main research axis developed over the last few years was to generate new pancreatic β-cells or to transplant pancreatic islets as occurring for some specific type 1 diabetes patients. We evaluate here a new paradigm consisting in preservation of β-cells by prevention of human islet amyloid polypeptide (hIAPP) oligomers and fibrils formation leading to pancreatic β-cell death. We review the hIAPP physiology and the pathology that contributes to β-cell destruction, deciphering the various cellular steps that could be involved. Recent progress in understanding other amyloidosis such as Aβ, Tau, α-synuclein or prion, involved in neurodegenerative processes linked with inflammation, has opened new research lines of investigations to preserve neuronal cells. We evaluate and estimate their transposition to the pancreatic β-cells preservation. Among them is the control of reactive oxygen species (ROS) production occurring with inflammation and the possible implication of the mitochondrial translocator protein as a diagnostic and therapeutic target. The present review also focuses on other amyloid forming proteins from molecular to physiological and physiopathological points of view that could help to better decipher hIAPP-induced β-cell death mechanisms and to prevent hIAPP fibril formation.
Collapse
Affiliation(s)
- Ghislaine Guillemain
- Sorbonne Université, Institut Hospitalo-Universitaire, Inserm UMR_S938, Institute of Cardio metabolism and Nutrition (ICAN), Centre de recherche de St-Antoine (CRSA), 27 rue de Chaligny, F-75012 Paris, France.
| | - Jean-Jacques Lacapere
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS UMR 7203, Laboratoire des BioMolécules (LBM), 4 place Jussieu, F-75005 Paris, France.
| | - Lucie Khemtemourian
- CBMN, CNRS UMR 5248, IPB, Univ. Bordeaux, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France.
| |
Collapse
|
6
|
Hu W, Dai CL, Niu J, Iqbal K. Heightened Tameness and Accelerated Handling-Habituation in 3×Tg-AD Mice on a B6;129 Genetic Background. J Alzheimers Dis Rep 2022; 6:245-255. [PMID: 35719714 PMCID: PMC9198767 DOI: 10.3233/adr-220007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/26/2022] [Indexed: 11/15/2022] Open
Abstract
Background: The triple transgenic mouse model of Alzheimer’s disease (3×Tg-AD) has gained popularity in Alzheimer’s research owing to the progressive development of both amyloid-β and tau pathologies in its brain. Prior handling-habituation, a necessary preparation procedure that reduces anxiety and stress in rodents, was seldom described in the literature involving these mice and needs to be addressed. Objective: We sought to determine whether 3×Tg-AD mice differ from B6;129 genetic control mice in terms of tameness and prior habituation to handling. Methods: We devised hand-staying and hand-boarding assays to evaluate tameness in 3×Tg-AD and B6;129 genetic control mice at 2.5, 7, and 11.5 months of age, representing cognitively pre-symptomatic, early symptomatic and advanced symptomatic stages of the disease, respectively. We monitored the progress of handling-habituation across 8–15 daily handling sessions and assessed the animal behaviors in elevated plus maze. Results: We found that 3×Tg-AD mice were markedly tamer than age-matched control mice at the baseline. Whereas it took 2–3 days for 3×Tg-AD mice to reach the criteria for full tameness, it took an average of 7–9 days for young genetic control mice to do so. Prior handling-habituation enhanced risk assessment and coping strategy in mice in elevated plus maze. Completely handling-habituated mice exhibited comparable anxiety indices in the maze regardless of genotype and age. Conclusion: These findings collectively point to inherently heightened tameness and accelerated handling-habituation in 3×Tg-AD mice on a B6;129 genetic background. These traits should be carefully considered when behaviors are compared between 3×Tg-AD and the genetic control mice.
Collapse
Affiliation(s)
- Wen Hu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Chun-Ling Dai
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Jiahui Niu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| |
Collapse
|
7
|
Sinsky J, Pichlerova K, Hanes J. Tau Protein Interaction Partners and Their Roles in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2021; 22:9207. [PMID: 34502116 PMCID: PMC8431036 DOI: 10.3390/ijms22179207] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Tau protein plays a critical role in the assembly, stabilization, and modulation of microtubules, which are important for the normal function of neurons and the brain. In diseased conditions, several pathological modifications of tau protein manifest. These changes lead to tau protein aggregation and the formation of paired helical filaments (PHF) and neurofibrillary tangles (NFT), which are common hallmarks of Alzheimer's disease and other tauopathies. The accumulation of PHFs and NFTs results in impairment of physiological functions, apoptosis, and neuronal loss, which is reflected as cognitive impairment, and in the late stages of the disease, leads to death. The causes of this pathological transformation of tau protein haven't been fully understood yet. In both physiological and pathological conditions, tau interacts with several proteins which maintain their proper function or can participate in their pathological modifications. Interaction partners of tau protein and associated molecular pathways can either initiate and drive the tau pathology or can act neuroprotective, by reducing pathological tau proteins or inflammation. In this review, we focus on the tau as a multifunctional protein and its known interacting partners active in regulations of different processes and the roles of these proteins in Alzheimer's disease and tauopathies.
Collapse
Affiliation(s)
| | | | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia; (J.S.); (K.P.)
| |
Collapse
|
8
|
Soni N, Medeiros R, Alateeq K, To XV, Nasrallah FA. Diffusion Tensor Imaging Detects Acute Pathology-Specific Changes in the P301L Tauopathy Mouse Model Following Traumatic Brain Injury. Front Neurosci 2021; 15:611451. [PMID: 33716645 PMCID: PMC7943881 DOI: 10.3389/fnins.2021.611451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/25/2021] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI) has been linked with tauopathy. However, imaging methods that can non-invasively detect tau-protein abnormalities following TBI need further investigation. This study aimed to investigate the potential of diffusion tensor imaging (DTI) to detect tauopathy following TBI in P301L mutant-tau-transgenic-pR5-mice. A total of 24 9-month-old pR5 mice were randomly assigned to sham and TBI groups. Controlled cortical injuries/craniotomies were performed for TBI/sham groups followed by DTI data acquisition on days 1 and 7 post-injury. DTI data were analyzed by using voxelwise analysis and track-based spatial statistics for gray matter and white matter. Further, immunohistochemistry was performed for total-tau and phosphorylated-tau, astrocytes, and microglia. To detect the association of DTI with these pathological markers, a correlation analysis was performed between DTI and histology findings. At day 1 post-TBI, DTI revealed a widespread reduction in fractional anisotropy (FA) and axial diffusivity (AxD) in the TBI group compared to shams. On day 7, further reduction in FA, AxD, and mean diffusivity and increased radial diffusivity were observed. FA was significantly increased in the amygdala and cortex. Correlation results showed that in the ipsilateral hemisphere FA reduction was associated with increased phosphorylated-tau and glial-immunoreactivity, whereas in the contralateral regions, the FA increase was associated with increased immunostaining for astrocytes. This study is the first to exploit DTI to investigate the effect of TBI in tau-transgenic mice. We show that alterations in the DTI signal were associated with glial activity following TBI and would most likely reflect changes that co-occur with/without phosphorylated-tau. In addition, FA may be a promising measure to identify discrete pathological processes such as increased astroglia activation, tau-hyperphosphorylation or both in the brain following TBI.
Collapse
Affiliation(s)
- Neha Soni
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Rodrigo Medeiros
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Khawlah Alateeq
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Xuan Vinh To
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Fatima A Nasrallah
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
9
|
An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. Neurosci Biobehav Rev 2020; 120:372-386. [PMID: 33171143 DOI: 10.1016/j.neubiorev.2020.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
L.P. Li, J.W. Liang and H.J. Fu. An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Traumatic brain injury (TBI) and Alzheimer's disease (AD) are devastating conditions that have long-term consequences on individual's cognitive functions. Although TBI has been considered a risk factor for the development of AD, the link between TBI and AD is still in debate. Aggregation of hyperphosphorylated tau and intercorrelated synaptic dysfunction, two key pathological elements in both TBI and AD, play a pivotal role in mediating neurodegeneration and cognitive deficits, providing a mechanistic link between these two diseases. In the first part of this review, we analyze the experimental literatures on tau pathology in various TBI models and review the distribution, biological features and mechanisms of tau pathology following TBI with implications in AD pathogenesis. In the second part, we review evidences of TBI-mediated structural and functional impairments in synapses, with a focus on the overlapped mechanisms underlying synaptic abnormalities in both TBI and AD. Finally, future perspectives are proposed for uncovering the complex relationship between TBI and neurodegeneration, and developing potential therapeutic avenues for alleviating cognitive deficits after TBI.
Collapse
|