-
Categories
-
Pharmaceutical Intermediates
-
Active Pharmaceutical Ingredients
-
Food Additives
- Industrial Coatings
- Agrochemicals
- Dyes and Pigments
- Surfactant
- Flavors and Fragrances
- Chemical Reagents
- Catalyst and Auxiliary
- Natural Products
- Inorganic Chemistry
-
Organic Chemistry
-
Biochemical Engineering
- Analytical Chemistry
-
Cosmetic Ingredient
- Water Treatment Chemical
-
Pharmaceutical Intermediates
Promotion
ECHEMI Mall
Wholesale
Weekly Price
Exhibition
News
-
Trade Service
Editor—Summer Leaf
of the CNS.
In the pathogenesis and disease progression of Alzheimer's disease (AD), this cross-dialogue between cells can amplify its function
through mutual crosstalk.
Therefore, when one of the cells has pathological changes, it often directly or indirectly affects other cells, and microglia play a leading role
in this process.
By elucidating existing drugs and therapeutics, targeting microglia and dialogue with other cells at different time windows may help alleviate neuroinflammation caused by AD, reduce neuronal death and provide an appropriate microenvironment
for nerve regeneration according to the characteristics of microglial continuity.
This may provide new ideas
for the treatment of neuroinflammatory degenerative diseases such as AD, which are currently progressing slowly.
Previous studies have used the two ends of microglia polarization as the basis for distinguishing the M1 pro-inflammatory/M2 anti-inflammatory paradigm, but this dichotomy is too simple to fully describe the dynamic change process
of microglial polarization.
Through literature research, it is found that distinguishing metabolic pathways such as microglia surface markers, cellular glucose metabolism, and cell epigenetic markers helps us understand the characteristics of
microglial continuity changes in different time windows in AD state.
Microglia usually accumulate around amyloid plaques and Tau protein due to their characteristic chemotaxis, and microglia have extensive dialogue and crosstalk
with other cells under normal physiological or AD pathological conditions.
This suggests that microglia play a key role in neuroinflammatory diseases such as AD, and by exploring the mechanism of interaction between microglia and other cells, it will provide a theoretical basis for reducing neuroinflammation and delaying neuroinflammatory degenerative diseases such as AD.
October 10, 2022, Ma Stub, Shanxi University of Traditional Chinese Medicine Based on the research results of his team and other teams in recent years, the professor's research group published a paper entitled "Neural Regeneration Research" Review article "The effects and potential of microglial polarization and crosstalk with other cells of the central nervous system in the treatment of Alzheimer's disease"
。 Master students Yige Wu and Associate Professor Lijuan Song are the co-first authors of the paper, and Professor Ma Xinggen is the corresponding author
.
The microglia phenotype in AD was analyzed to be characterized by continuous changes and extensive dialogue
with other CNS innate cells and peripherally invading immune cells.
Patients with the disease may benefit
from targeting microglial phenotypic changes and dialogue with other cells.
(Read more: The latest progress of the Ma stub team, see Logic Neuroscience report: Front Cell Neurosci.
) - Using bibliometrics to analyze the research trends of astrocytes and stroke; Neural Regen Res Review—Advantages of Rho kinase and its inhibitor Fasduil in the treatment of neurodegenerative diseases; A Review of Front Aging Neurosci – The double-edged role of astrocytes in neurovascular units after cerebral ischemia)
Microglia are disease regulators in AD
AD is a degenerative disease of CNS, with cognitive function and behavioral disorders as the main clinical features, and has received increasing attention in modern society [1].
As innate immune cells of the CNS, microglia can participate in the occurrence and progression of neurodegeneration by activating inflammatory signaling pathways, thereby affecting the prognosis of AD patients [2].
Based on specific characteristics, activated microglia can be divided into two broad categories: M1 and M2 [3-9].
This simplified dichotomy is still used in clinical and experimental research
.
However, as far as current research progress is concerned[10-12], microglia in AD are known to exist in a number of different activation states, and the continuity of microglial polarization can be demonstrated by describing their continuity changes (Figure 1).
。 Currently, polarization of pro-inflammatory M1 microglia is generally thought to be associated with chronic neuroinflammation in AD, with the deposition of β-amyloid (Aβ) follows CNS inflammation levels increase [13].
Even the many factors produced by M2 type microglia, which are thought to have anti-inflammatory functions, are not entirely beneficial to AD [14].
Therefore, in order to treat AD, it is necessary to adjust the delicate therapeutic balance
by altering the polarization of microglia at different stages of AD.
Figure 1 The continuous process of microglial polarization (Source: Wu Y, et al.
, Neural Regen Res, 2022).
Migration and phagocytosis of AD microglia
Microglia, as the main immune cells responsible for maintaining homeostasis within the CNS, continuously monitor for signs of neurological damage (such as pathogen invasion or tissue damage) and then generate a cascade of responses to address damaging factors encountered in immune defenses [15.
16]
。 During the pathogenesis of AD, microglia are constantly activated and subsequently become round migrating cells called "amoeba" cells [17,18].
These activated microglia exhibit migration and phagocytosis capabilities, encapsulating Aβ and degrading other substances [19], which in turn affects the prognosis
of AD.
(1) Migration of microglia in AD
The formation of amyloid plaques and tau protein tangles in the brain is a typical pathological feature of AD, and microglia usually gather around them due to the ability of amyloid plaques and tau proteins in AD patients to migrate [20]。 One of the main processes by which microglia transition from a quiescent to an activated state is migration and aggregation [21].
However, the specific mechanism behind the migration remains unknown
.
The migration effect of microglia is related to the release of multiple cytokines and the activation of many signaling pathways [22].
Microglia reside in the CNS in a physiological state and are called resting microglia
.
They have the characteristics of small cytoplasms, elongated protrusions and more branches on the surface, and are the gatekeepers of the CNS [23].
Microglia are constantly moving, can sense changes in the surrounding environment, and maintain the body's homeostasis
.
However, microglia in pathological states such as AD are activated by a variety of factors and surface-related receptors, such as TREM2 and toll-like receptors (TLRs), which can be associated with Aβ and apolipoprotein E (ApoE) bind to each other and migrate to the site of injury [24,25].
(2) Phagocytosis of microglia in AD
During the pathogenesis of AD, Aβ deposition in the brain is often accompanied by glial cell proliferation and lipid deposition, which leads to a continuous progression of the inflammatory response [26].In addition, microglia can recognize amyloid through receptors such as TREM2, TLRs, and CR1, creating a unique signaling cascade
.
This process drives microglia recognition and migration to the lesion site [27].
Subsequently, microglia begin to engulf age age spots (SPs) and other pathogenic products by phagocytosis and microphagocytosis [28,29].
Phagocytosis allows soluble Aβ and tau proteins to be absorbed into microglia, and upon binding to intracellular substances and fusion with endosomes and lysosomes, vesicles and their contents are destroyed by acid hydrolases and tissue hydrolases in lysosomes, anti-inflammatory mediators are released, and lipid clearance is improved[30]
。
Microglia can directly engulf AD toxic products such as Aβ and tau proteins, which have advantages
in AD therapy.
However, recent studies have shown that microglia phagocytosis is not always beneficial, in part related to specific clinical stages [31].
In patients with only pathophysiological changes in AD but no or only mild clinical symptoms, enhanced microglial phagocytosis reduces Aβ levels, slows the formation of SP, and prevents the production of nerve fiber tangles (NFTs), These help prevent the onset of AD and delay the clinical progression of AD [32].
Due to the absence of lysosomes, the phagocytic capacity of microglia decreases after phagocytosis of Aβ and tau proteins [33].
What's more, after exerting phagocytosis, the cell metabolism produces a cascade of inflammatory vesicles (such as NLRP3) and inflammatory cytokines (such as TNF-α, IL-1) The presence of microglia in turn causes microglia to polarize in the pro-inflammatory direction, thereby inducing the secretion of pro-inflammatory cytokines, exacerbating the inflammatory response, reducing the phagocytosis of Aβ, and promoting the pathological response to tau protein [34].
At the same time, microglia's phagocytic capacity decreases with age or prolonged exposure to increased Aβ load [35].
Crosstalk between microglia and other cells in AD
Different types of glial cells in the CNS, including microglia, astrocytes, and oligodendrocytes, perform different functions and coordinate the homeostasis of the CNS and neurons through a variety of intercellular crosstalk mechanisms known as neuroimmunity [36].
Cells have the ability to amplify their function by crosstalk with each other [37].
Therefore, when diseases affect one of these cells, they usually directly or indirectly affect the other cells (Figure 2).
Figure 2 Crosstalk between microglia and other cells in Alzheimer's disease
(Source: Wu Y, et al.
, Neural Regen Res, 2022).
(1) Crosstalk between microglia and astrocytes in AD
Activation and polarization of microglia and astrocytes are often thought to exacerbate AD development and aging.
Although the underlying cellular and molecular mechanisms are unknown, multiple cytokines and signaling pathways are known to be involved in the activation
of microglia and astrocytes in AD.
In addition, activation and polarization of microglia often contribute to astrocyte activation, leading to neuronal toxicity and aggravating the inflammatory environment [38].
Similarly, due to astrocyte activation, more microglia transition from quiescent to activated
.
In AD, there is extensive dialogue and crosstalk between these two types of glial cells [39].
Polarized microglia communicate with astrocytes through inflammatory crosstalk and other mechanisms, activating and polarizing astrocytes [40].
Pathological products of AD (such as Aβ and tau) contribute to microglial polarization, predominate the pro-inflammatory phenotype of microglia, and induce pro-inflammatory factors such as IL-1β , IL-6, TNF-ɑ, and NO secretion [41].
These factors not only reduce the phagocytosis of microglia, but also trigger astrocytes to form a pro-inflammatory phenotype and increase the expression of pro-inflammatory factors [42].
Similarly, astrocyte activation contributes to microglial polarization
in AD.
When pathological products such as Aβ are stimulated, the NF-κB-related signaling pathway in astrocytes is activated, resulting in the release
of complement C3.
This activates C3 receptors on the surface of microglia and leads to impaired pro-inflammatory polarization and phagocytosis [43].
In short, inflammatory crosstalk between microglia and astrocytes occurs during the development and development of AD [44].
(2) Crosstalk between microglia and oligodendrocytes in AD
Oligodendrocytes, which play a key role in maintaining the function and integrity of axons and myelin, are also thought to be involved in AD [45].In a physiological state, oligodendrocytes are closely related to the production of steroids, substances that help protect neurons from damage caused by degenerative diseases [46].
Due to the neuroanti-inflammatory effect of steroids, the interaction between microglia and oligodendrocytes reduces neuroinflammation and prevents pro-inflammatory polarization
of microglia.
Similarly, increasing oligodendrocyte differentiation upregulates IGF-1 production [47], which is beneficial
for AD patients.
Although there are no experimental results to support the possible direct damage caused by Aβ or tau proteins to oligodendrocytes [48], some researchers have found too much Aβ or ApoE causes loss of myelin sheath integrity and induces oligodendrocyte apoptosis [49].
Previous studies have shown that oligodendrocytes may play an important role in the accumulation of tau protein in AD [50].
Single-cell sequencing techniques in the brains of AD patients reveal that oligodendrocytes exhibit the most individual variation among major brain cell types [51].
In addition, it has been hypothesized that inflammation reduces the ability of oligodendrocytes to form myelin [52].
These findings may shed light on new ideas for treating AD by interfering with communication between microglia and oligodendrocytes.
Since there has been a focus on the function of oligodendrocytes in maintaining myelin sheath integrity in the past, many studies have pointed to neuroinflammation-oriented diseases such as multiple sclerosis (MS).
Oligodendrocytes in AD are only seen as potential neuroprotective targets, so research into crosstalk dialogue between microglia and oligodendrocytes has just begun
.
(3) Crosstalk between AD microglia and neurons
The interaction between microglia and neurons plays an important role in the immune response throughout the development of AD [53].In a healthy brain, many molecules mediate two-way communication
between microglia and neurons.
On the one hand, neurons affect the function of microglia through neurotransmitters such as glutamic acid, dopamine, γ-aminobutyric acid (GABA), ADP, adenosine, and migration factors [54].
On the other hand, cytokines secreted by microglia such as GDNF and IGF-1 are also beneficial to maintain the physiological functions of neurons and play a key role in learning and memory [55].
。
Glutamate is the most important excitatory neurotransmitter in the CNS and is directly involved in crosstalk between microglia and neurons, and too much or too little glutamate release can have serious consequences for neurotransmission, leading to the development of AD [56].
However, under neuroinflammation and AD conditions, microglia tend to release excess glutamate, leading to neuronal excitotoxicity in AD and exacerbating neurodegenerative processes [57].
Cystine (Cys)/glutamate exchanger (Xc(-) exchanger), and d-serine release when stimulated by inflammation, directly affect the CNS glutamate levels in [58], which in turn affects the dialogue
between microglia and neurons.
Under conditions where AD persists chronic inflammation, the release of inflammatory factors such as TNF-α, IL-6, NO, and ROS increases neuronal death [59]
。 At the same time, the loss of homeostasis within microglia induces further release of pro-inflammatory factors and more neuronal death, leading to a vicious cycle [60].
As AD progresses, neurons are damaged
by an inflammatory environment induced by pro-inflammatory polarized microglia.
Due to the accumulation of Aβ, neurons themselves are induced to exhibit various pathological features [61]
.
Weakening of neuronal function, which in turn has a significant effect
on microglial polarization.
The interaction between microglia and neurons is like the two ends of a balancing beam, and damage at one end can wreak havoc on
the other.
The activation of microglia is not a single pro-inflammatory or anti-inflammatory polarization, but a series of changes such as differences in cell states or overlapping functions [62].
This may explain why as AD progresses, pro-inflammatory microglia increase, causing more neuronal death, leading to progressive clinical worsening
in AD patients.
(4) Crosstalk between microglia and peripheral innate immune cells in AD
There is growing evidence that peripheral immune cells enter the CNS through the damaged blood-brain barrier and then release cytokines such as IL-6, IL-1, and TNF-α [63]。 For example, T lymphocytes that migrate to the brains of AD patients are able to respond to Aβ [64].
In addition, various types of Aβ-specific T cells may promote enhanced Aβ clearance and weaken the inflammatory environment of the CNS, even reverse declining cognitive abilities [65]
。 In addition, in AD, specific crosstalk with microglia by peripherally recruited helper T lymphocytes by the production of the effector cytokine IFN-γ and activation of macrophages and CD8+ T cells to stimulate cells to respond
to harmful stimuli.
Currently, the specific mechanism of T helper lymphocyte function is unknown, although some researchers believe that in AD, helper T lymphocytes produce IFN-γ Helps microglia surround Aβ plaques and accelerate clearance [66].
However, it has also been suggested that T helper lymphocytes play an invasive role in AD, which leads to negative consequences through the common leukocyte recruitment process [67].
The exact role of cytotoxic T lymphocytes in the development of AD is unknown, but the infiltration observed in AD patients may indicate the involvement of cytotoxic T cells [68]
。 Thus, cytotoxic T cells and helper T lymphocytes appear to drive the development of AD and cause extra through crosstalk with microglia in chronic inflammation of AD CNS damage
.
IV.
Summary and Prospect
The results show that the dialogue between microglia and other cells in AD plays a key role
in CNS inflammation.
The dominant role of microglia in the inflammatory environment of AD and their interaction with other cells are reviewed
.
By exploring the mechanism of interaction between microglia and other cells, it provides a theoretical basis
for reducing neuroinflammation.
Reducing neuroinflammation is undoubtedly of great significance
for further research on nerve regeneration.
By regulating crosstalk between microglia and other cells, it may be possible to reduce neuronal death caused by inflammatory damage, while also providing a good environment
for the subsequent regeneration of new neurons.
In addition, since AD is often accompanied by disruption of the blood-brain barrier, it is difficult to predict crosstalk between innate cells and peripheral cells within the CNS, and whether these cells are beneficial to the treatment of AD remains controversial
.
The authors believe this review can provide researchers with new concepts
.
As Frozza et al.
point out, small advances in AD treatment strategies, even with a slight delay in the onset of onset, can reduce the overall burden of
disease.
Delaying the progression of AD by intervening in the polarization of microglia may pave the way for successful treatment of AD in the future
.
Original link: https://doi.
org/10.
4103/1673-5374.
355747
Funds: National Natural Science Foundation of China (81473577, 82004028), China Postdoctoral Science Foundation (2020M680912), Shanxi Provincial Applied Basic Research Program ( 201901D211538), Shanxi Provincial Department of Education Science and Technology Innovation Project for Colleges and Universities (2019L0734), Shanxi Provincial Health Commission Medical Science and Technology Leading Team (2020TD05), Shanxi Provincial Health Commission Key Laboratory of Neurological Disease Prevention and Control Research ( 2020SYS20), Shanxi University of Chinese Medicine Young Scientist Training Project (2021PY-QN-09) and Shanxi University of Traditional Chinese Medicine Discipline Construction Funding Project
.
First author: Master Wu Yige (left), Associate Professor Song Lijuan (middle), Corresponding author: Professor Ma Xinggen (right)
(Photo courtesy of: Neurobiology Research Center, Shanxi University of Chinese Medicine/Key Laboratory of Multiple Sclerosis and Blood Activation of the State Administration of Traditional Chinese Medicine)
Corresponding author and lab profile (swipe up and down to read).
Ma Xingen, Director of the Center/Research Office, Second-level Professor, PhD Supervisor, Subject Leader, Member of the Teaching Steering Committee of Traditional Chinese Medicine of the Ministry of Education; Vice Chairman of Zhongjing Inheritance and Innovation Professional Committee of the World Federation of Chinese Medicine Societies/Vice Chairman
of Zhongjing Academic Inheritance and Innovation Community of Chinese Society of Traditional Chinese Medicine.
His research direction is the prevention and treatment of central nervous system diseases
by combining traditional Chinese medicine and western medicine.
He has published more than 280 academic papers, including 212 Chinese core (or SCI) papers, and has been published by Immunol Res and Glia and other journal papers have been positively cited more than 2,000 times
.
The Neurobiology Research Center of Shanxi University of Chinese Medicine/Key Laboratory of Multiple Sclerosis and Qi and Blood Activation of the State Administration of Traditional Chinese Medicine is the key discipline
of the State Administration of Traditional Chinese Medicine 。 After years of construction, the discipline has gradually condensed and formed four stable research directions, namely the role and mechanism of combined traditional Chinese medicine and western medicine in the prevention and treatment of ischemic cerebrovascular diseases, the role and mechanism of integrated traditional Chinese and western medicine in the prevention and treatment of multiple sclerosis and other demyelinating diseases, the role and mechanism of integrated traditional Chinese medicine and western medicine in the prevention and treatment of Alzheimer's disease and vascular dementia, and the role and mechanism of
integrated traditional Chinese medicine in the prevention and treatment of Parkinson's disease and other movement disorders 。 These four directions are characterized by the prevention and treatment of the above diseases by the theory of traditional Chinese medicine.
Based on the pathological characteristics of neuroinflammation, oxidative stress and abnormal immune response as the basis of co-pathogenesis, the research advantage
of using the traditional theory of traditional Chinese medicine "treating different diseases together" has been formed.
[1] Mol Neurobiol—Zhu Lixin/Guo Jiasong's team revealed the important role of the basement membrane protein Perlecan in the recovery of blood-spinal cord barrier function after spinal cord injury
[2] Transl Psychiatry—Yang Xun's team revealed the different neural mechanisms of schizophrenia in the reward expectation and reward outcome stages
[3] Schizophrenia—Brain network hub center and working memory performance in patients with schizophrenia
[4] Front Cell Neurosci—Using bibliometrics to analyze research trends in astrocytes and stroke
[5] Cereb Cortex-Liu Tao's team revealed the accelerated degradation pattern of white matter structure in the elderly
[6] Biophys J—Professor Xu Guangkui's research group reveals the network dynamics of nonlinear power-law relaxation in the cell cortex
[7] Science—Analysis of neurogenesis and regeneration in Mexican axolotls using single-cellular, multi-omics techniques
[8] J Neuroinflammation Review—Ni Wenfei/Zhou Kailiang's team focused on the important role of STING pathway in neuroinflammation and cell death after CNS injury
[9] Sci Adv-Sheng Neng-Yin/Mao Bingyu/Ding Yuqiang teamed up to discover a new mechanism of AMPA receptor ubiquitination in the regulation of excitatory synaptic function
[10] Brain—Liu Gang/Hu Qingmao's team revealed the driving role of auxiliary motor areas in the alteration of the whole brain structural network in patients with blepharospasm
Recommended high-quality scientific research training courses[1] Symposium on Single Cell Sequencing and Spatial Transcriptomics Data Analysis (October 29-30 Tencent Online Conference) Conference/Forum/Seminar Preview[1] Immune Zoom Seminar—Screening of B cells in the immune and nervous system (Professor Xu Heping)
[2] Academic Conference - 2022 Symposium on Neural Circuit Tracing Technology and the Second Round of the Second Round of the 6th National Training Course on Neural Circuit Tracing Technology
[3] Roundtable – Xu Fuqiang/Jia Yichang/Han Lanqing/Cai Lei et al.
discussed gene therapy innovation for neurodegenerative diseases and ophthalmic diseases
Reference (Swipe up and down to read).
[1] Tublin JM, Adelstein JM, Del Monte F, Combs CK, Wold LE (2019) Getting to the heart of Alzheimer disease.
Circ Res 124:142-149.
[2] Doust YV, King AE, Ziebell JM (2021) Implications for microglial sex differences in tau-related neurodegenerative diseases.
Neurobiol Aging 105:340-348.
[3] Franco R, Lillo A, Rivas-Santisteban R, Reyes-Resina I, Navarro G (2021) Microglial adenosine receptors: from preconditioning to modulating the M1/M2 balance in activated cells.
Cells 10:1124.
[4] Ransohoff RM (2016) A polarizing question: do M1 and M2 microglia exist? Nat Neurosci 19:987-991.
[5] Devanney NA, Stewart AN, Gensel JC (2020) Microglia and macrophage metabolism in CNS injury and disease: The role of immunometabolism in neurodegeneration and neurotrauma.
Exp Neurol 329:113310.
[6] Wright-Jin EC, Gutmann DH (2019) Microglia as dynamic cellular mediators of brain function.
Trends Mol Med 25:967-979.
[7] Cornell J, Salinas S, Huang HY, Zhou M (2022) Microglia regulation of synaptic plasticity and learning and memory.
Neural Regen Res 17(4):705-716.
[8] Chhor V, Le Charpentier T, Lebon S, Oré MV, Celador IL, Josserand J, Degos V, Jacotot E, Hagberg H, Sävman K, Mallard C, Gressens P, Fleiss B.
Characterization of phenotype markers and neuronotoxic potential of polarised primary microglia in vitro (2013) Brain Behav Immun 32:70-85.
[9] Sarlus H, Heneka MT (2017) Microglia in Alzheimer's disease 127:3240-3249.
[10] Yao K, Zu HB (2019) Microglial polarization: novel therapeutic mechanism against Alzheimer's disease.
Inflammopharmacology 28:95-110.
[11] Koh YC, Yang G, Lai CS, Weerawatanakorn M, Pan MH (2018) Chemopreventive effects of phytochemicals and medicines on M1/M2 polarized macrophage role in inflammation-related diseases.
Int J Mol Sci 19:2208.
[12] McQuade A, Blurton-Jones M (2019) Microglia in Alzheimer’s disease: exploring how genetics and phenotype influence risk.
J Mol Biol 431:1805-1817.
[13] Ozben T, Ozben S (2019) Neuro-inflammation and anti-inflammatory treatment options for Alzheimer's disease.
Clin Biochem 72:87-89.
[14] Guillot-Sestier MV, Doty KR, Gate D, Rodriguez J Jr, Leung BP, Rezai-Zadeh K, Town T (2015) Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology.
Neuron 85:534-548.
[15] Weinhard L, di Bartolomei G, Bolasco G, Machado P, Schieber NL, Neniskyte U, Exiga M, Vadisiute A, Raggioli A, Schertel A, Schwab Y, Gross CT (2018) Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction.
Nat Commun 9:1228.
[16] Li Q, Barres BA (2018) Microglia and macrophages in brain homeostasis and disease.
Nat Rev Immunol 18:225-242.
[17] Lau SF, Fu AKY, Ip NY (2021) Cytokine signaling convergence regulates the microglial state transition in Alzheimer's disease.
Cell Mol Life Sci 78:4703-4712.
[18] Das R, Chinnathambi S (2020) Actin-mediated microglial chemotaxis via g-protein coupled purinergic receptor in Alzheimer’s disease.
Neuroscience 448:325-336.
[19] Huang Y, Happonen KE, Burrola PG, O'Connor C, Hah N, Huang L, Nimmerjahn A, Lemke G (2021) Microglia use TAM receptors to detect and engulf amyloid β plaques.
Nat Immunol 22:586-594.
[20] Zhang X, Ye P, Wang D, Liu Y, Cao L, Wang Y, Xu Y, Zhu C (2019) Involvement of rhoa/rock signaling in aβ-induced chemotaxis, cytotoxicity and inflammatory response of microglial BV2 cells.
Cell Mol Neurobiol 39:637-650.
[21] Yu T, Lin Y, Xu Y, Dou Y, Wang F, Quan H, Zhao Y, Liu X (2020) Repressor element 1 silencing transcription factor (REST) governs microglia-like BV2 cell migration via progranulin (PGRN).
Neural Plast 2020:8855822.
[22] Chidambaram H, Das R, Chinnathambi S (2022) G-protein coupled purinergic P2Y12 receptor interacts and internalizes TauRD-mediated by membrane-associated actin cytoskeleton remodeling in microglia.
Eur J Cell Biol 101:151201.
[23] Kreutzberg GW (1996) Microglia: a sensor for pathological events in the CNS.
Trends Neurosci 19:312-318.
[24] Ulrich JD, Ulland TK, Mahan TE, Nyström S, Nilsson KP, Song WM, Zhou Y, Reinartz M, Choi S, Jiang H, Stewart FR, Anderson E, Wang Y, Colonna M, Holtzman DM (2018) ApoE facilitates the microglial response to amyloid plaque pathology.
J Exp Med 215:1047-1058.
[25] Salcman B, Affleck K, Bulfone-Paus S (2021) P2X receptor-dependent modulation of mast cell and glial cell activities in neuroinflammation.
Cells 10:2282.
[26] Domingues C, da Cruz E Silva OAB, Henriques AG (2017) Impact of cytokines and chemokines on Alzheimer’s disease neuropathological hallmarks.
Curr Alzheimer Res 14:870-882.
[27] Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, Itzkovitz S, Colonna M, Schwartz M, Amit I (2017) A unique microglia type associated with restricting development of Alzheimer’s disease.
Cell 169:1276-1290.
e17.
[28] Podleśny-Drabiniok A, Marcora E, Goate AM (2020) Microglial phagocytosis: a disease-associated process emerging from Alzheimer’s disease genetics.
Trends Neurosci 43:965-979
[29] Bolmont T, Haiss F, Eicke D, Radde R, Mathis CA, Klunk WE, Kohsaka S, Jucker M, Calhoun ME (2008) Dynamics of the microglial/amyloid interaction indicate a role in plaque maintenance.
J Neurosci 28:4283-92.
[30] Doran AC, Yurdagul A Jr, Tabas I (2020) Efferocytosis in health and disease.
Nat Rev Immunol 20:254-267.
[31] Anwar S, Rivest S (2020) Alzheimer's disease: microglia targets and their modulation to promote amyloid phagocytosis and mitigate neuroinflammation.
Expert Opin Ther Targets 24:331-344.
[32] Wang Y, Ulland TK, Ulrich JD, Song W, Tzaferis JA, Hole JT, Yuan P, Mahan TE, Shi Y, Gilfillan S, Cella M, Grutzendler J, DeMattos RB, Cirrito JR, Holtzman DM, Colonna M (2016) TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques.
J Exp Med 213:667-675.
[33] Carosi JM, Sargeant TJ (2019) Rapamycin and Alzheimer disease: a double-edged sword?.
Autophagy15:1460-1462.
[34] van Olst L, Verhaege D, Franssen M, Kamermans A, Roucourt B, Carmans S, Ytebrouck E, van der Pol SMA, Wever D, Popovic M, Vandenbroucke RE, Sobrino T, Schouten M, de Vries HE (2020) Microglial activation arises after aggregation of phosphorylated-tau in a neuron-specific P301S tauopathy mouse model.
Neurobiol Aging 89:89-98.
[35] Rajendran L, Paolicelli RC (2018) Microglia-mediated synapse loss in Alzheimer’s disease.
J Neurosci 38:2911-2919.
[36] Delpech JC, Herron S, Botros MB, Ikezu T (2019) Neuroimmune crosstalk through extracellular vesicles in health and disease.
Trends Neurosci 42:361-372.
[37] Lana D, Ugolini F, Nosi D, Wenk GL, Giovannini MG (2021) The emerging role of the interplay among astrocytes, microglia, and neurons in the hippocampus in health and disease.
Front Aging Neurosci 13:651973.
[38] Fakhoury M (2018) Microglia and Astrocytes in Alzheimer’s disease: implications for therapy.
Curr Neuropharmacol 16:508-518.
[39] Guttikonda SR, Sikkema L, Tchieu J, Saurat N, Walsh RM, Harschnitz O, Ciceri G, Sneeboer M, Mazutis L, Setty M, Zumbo P, Betel D, de Witte LD, Pe'er D, Studer L (2021) Fully defined human pluripotent stem cell-derived microglia and tri-culture system model C3 production in Alzheimer's disease.
Nat Neurosci 24:343-354.
[40] Xie L, Zhang N, Zhang Q, Li C, Sandhu AF, Iii GW, Lin S, Lv P, Liu Y, Wu Q, Yu S (2020) Inflammatory factors and amyloid β-induced microglial polarization promote inflammatory crosstalk with astrocytes.
Aging (Albany NY) 12:22538-22549.
[41] Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M (2019) Alzheimer's disease: pathogenesis, diagnostics, and therapeutics.
Int J Nanomedicine 14:5541-5554.
[42] Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Münch AE, Chung WS, Peterson TC, Wilton DK, Frouin A, Napier BA, Panicker N, Kumar M, Buckwalter MS, Rowitch DH, Dawson VL, Dawson TM, Stevens B, et al.
(2017) Neurotoxic reactive astrocytes are induced by activated microglia.
Nature 541:481-487.
[43] Lian H, Litvinchuk A, Chiang AC, Aithmitti N, Jankowsky JL, Zheng H (2016) Astrocyte-microglia cross talk through complement activation modulates amyloid pathology in mouse models of Alzheimer’s disease.
J Neurosci 36:577-89.
[44] Rostami J, Mothes T, Kolahdouzan M, Eriksson O, Moslem M, Bergström J, Ingelsson M, O'Callaghan P, Healy LM, Falk A, Erlandsson A (2021) Crosstalk between astrocytes and microglia results in increased degradation of α-synuclein and amyloid-β aggregates.
J Neuroinflammation 18:124.
[45] Skaper SD, Facci L, Zusso M, Giusti P (2018) An inflammation-centric view of neurological disease: beyond the neuron.
Front Cell Neurosci 12:72.
[46] Chavda V, Singh K, Patel V, Mishra M, Mishra AK (2022) Neuronal glial crosstalk: specific and shared mechanisms in Alzheimer’s disease.
Brain Sci 12:75.
[47] Kabir MT, Uddin MS, Zaman S, Rahman MS, Behl T, Ahmad A, Hafeez A, Perveen A, Ashraf GM (2021) Exploring the anti-neuroinflammatory potential of steroid and terpenoid-derived phytochemicals to combat Alzheimer’s disease.
Curr Pharm Des 27:2635-2647.
[48] Chen S, Wang T, Yao J, Brinton RD (2020) Allopregnanolone promotes neuronal and oligodendrocyte differentiation in vitro and in vivo: therapeutic implication for Alzheimer's disease.
Neurotherapeutics 17:1813-1824.
[49] Li L, Li R, Zacharek A, Wang F, Landschoot-Ward J, Chopp M, Chen J, Cui X (2020) ABCA1/ApoE/HDL signaling pathway facilitates myelination and oligodendrogenesis after stroke.
Int J Mol Sci 21:4369.
[50] Nasrabady SE, Rizvi B, Goldman JE, Brickman AM (2018) White matter changes in Alzheimer's disease: a focus on myelin and oligodendrocytes.
Acta Neuropathol Commun 6:22.
[51] Ferrer I, Aguiló García M, Carmona M, Andrés-Benito P, Torrejón-Escribano B, Garcia-Esparcia P, Del Rio JA (2019) Involvement of oligodendrocytes in tau seeding and spreading in tauopathies.
Front Aging Neurosci 11:112.
[52] Grubman A, Chew G, Ouyang JF, Sun G, Choo XY, McLean C, Simmons RK, Buckberry S, Vargas-Landin DB, Poppe D, Pflueger J, Lister R, Rackham OJL, Petretto E, Polo JM (2019) A single-cell atlas of entorhinal cortex from individuals with Alzheimer's disease reveals cell-type-specific gene expression regulation.
Nat Neurosci 22(:2087-2097.
[53] Lorenzini L, Fernandez M, Baldassarro VA, Bighinati A, Giuliani A, Calzà L, Giardino L (2020) White matter and neuroprotection in Alzheimer's dementia.
Molecules 25:503.
[54] Tajbakhsh A, Read M, Barreto GE, Ávila-Rodriguez M, Gheibi-Hayat SM, Sahebkar A (2021) Apoptotic neurons and amyloid-beta clearance by phagocytosis in Alzheimer's disease: pathological mechanisms and therapeutic outlooks.
Eur J Pharmacol 895:173873.
[55] Czapski GA, Strosznajder JB (2021) Glutamate and GABA in microglia-neuron cross-talk in Alzheimer's disease.
Int J Mol Sci 22:11677.
[56] Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ, Hempstead BL, Littman DR, Gan WB (2013) Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor.
Cell 155:1596-609.
[57] Bukke VN, Archana M, Villani R, Romano AD, Wawrzyniak A, Balawender K, Orkisz S, Beggiato S, Serviddio G, Cassano T (2020) The dual role of glutamatergic neurotransmission in Alzheimer's disease: from pathophysiology to pharmacotherapy.
Int J Mol Sci 21:7452.
[58] Wang R, Reddy PH (2017) Role of glutamate and NMDA receptors in Alzheimer’s disease.
J Alzheimers Dis 57:1041-1048.
[59] Henstridge CM, Hyman BT, Spires-Jones TL (2019) Beyond the neuron-cellular interactions early in Alzheimer disease pathogenesis.
Nat Rev Neurosci 20:94-108.
[60] Spangenberg EE, Green KN (2017) Inflammation in Alzheimer's disease: Lessons learned from microglia-depletion models.
Brain Behav Immun 61:1-11.
[61] De Strooper B, Karran E (2016) The cellular phase of Alzheimer’s disease.
Cell 164:603-615.
[62] Merlo S, Spampinato SF, Caruso GI, Sortino MA (2020) The ambiguous role of microglia in Aβ toxicity: chances for therapeutic intervention.
Curr Neuropharmacol 18:446-455.
[63] Laurent C, Dorothée G, Hunot S, Martin E, Monnet Y, Duchamp M, Dong Y, Légeron FP, Leboucher A, Burnouf S, Faivre E, Carvalho K, Caillierez R, Zommer N, Demeyer D, Jouy N, Sazdovitch V, Schraen-Maschke S, Delarasse C, Buée L, et al.
(2016) Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy.
Brain 140:184-200.
[64] Wu Q, Kong W, Wang S (2021) Peripheral blood biomarkers CXCL12 and TNFRSF13C associate with cerebrospinal fluid biomarkers and infiltrating immune cells in Alzheimer disease.
J Mol Neurosci 71:1485-1494.
[65] Vacinova G, Vejražkova D, Rusina R, Holmerová I, Vaňková H, Jarolímová E, Včelák J, Bendlová B, Vaňková M (2021) Regulated upon activation, normal T cell expressed and secreted (RANTES) levels in the peripheral blood of patients with Alzheimer's disease.
Neural Regen Res 16:796-800.
[66] Fisher Y, Nemirovsky A, Baron R, Monsonego A (2010) T cells specifically targeted to amyloid plaques enhance plaque clearance in a mouse model of Alzheimer's disease.
PLoS One 5:e10830.
[67] Gate D, Saligrama N, Leventhal O, Yang AC, Unger MS, Middeldorp J, Chen K, Lehallier B, Channappa D, De Los Santos MB, McBride A, Pluvinage J, Elahi F, Tam GK, Kim Y, Greicius M, Wagner AD, Aigner L, Galasko DR, Davis MM, et al.
(2020) Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer's disease.
Nature 577:399-404.
[68] Unger MS, Li E, Scharnagl L, Poupardin R, Altendorfer B, Mrowetz H, Hutter-Paier B, Weiger TM, Heneka MT, Attems J, Aigner L (2020) CD8+ T-cells infiltrate Alzheimer's disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice.
Brain Behav Immun 89:67-86.
End of this article