Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2149 2024-02-08 09:58:10 |
2 layout Meta information modification 2149 2024-02-08 10:12:45 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Avola, R.; Furnari, A.G.; Graziano, A.C.E.; Russo, A.; Cardile, V. Neuroinflammation in Neurodegenerative Diseases. Encyclopedia. Available online: https://encyclopedia.pub/entry/54930 (accessed on 05 July 2024).
Avola R, Furnari AG, Graziano ACE, Russo A, Cardile V. Neuroinflammation in Neurodegenerative Diseases. Encyclopedia. Available at: https://encyclopedia.pub/entry/54930. Accessed July 05, 2024.
Avola, Rosanna, Alessandro Giuseppe Furnari, Adriana Carol Eleonora Graziano, Alessandra Russo, Venera Cardile. "Neuroinflammation in Neurodegenerative Diseases" Encyclopedia, https://encyclopedia.pub/entry/54930 (accessed July 05, 2024).
Avola, R., Furnari, A.G., Graziano, A.C.E., Russo, A., & Cardile, V. (2024, February 08). Neuroinflammation in Neurodegenerative Diseases. In Encyclopedia. https://encyclopedia.pub/entry/54930
Avola, Rosanna, et al. "Neuroinflammation in Neurodegenerative Diseases." Encyclopedia. Web. 08 February, 2024.
Neuroinflammation in Neurodegenerative Diseases
Edit

Neuroinflammation is a complex innate immune response occurring in the central nervous system (CNS), orchestrated primarily by specialized resident cells, notably glial cells, with microglia and astrocytes taking center stage.

neuroinflammation oxidative stress essential oils natural compounds

1. Introduction

Neuroinflammation is a complex innate immune response occurring in the central nervous system (CNS), orchestrated primarily by specialized resident cells, notably glial cells, with microglia and astrocytes taking center stage. Pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) trigger neuroinflammation as they are recognized by pattern recognition receptors (PRRs) expressed in microglia, which, as a consequence, activate their phagocytic capabilities and the release of signaling molecules that mediate the neuroinflammatory response by allowing the activation and recruitment of other immune cells in situ. This physiological immune process aims to protect against pathogens or damaged cells. However, when uncontrolled and prolonged, it can lead to neuronal death and neurodegeneration.
To degrade phagocytosed material, activated microglia generate neurotoxic reactive oxygen species (ROS) and reactive nitrogen species (RNS), resulting in harmful effects on neural tissue. In addition, pro-inflammatory molecules might stimulate neuronal cell death and increase blood–brain barrier permeability, disrupting its integrity [1][2][3]. Neuroinflammation is a common feature of many brain diseases such as Alzheimer’s disease, Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), frontotemporal lobar dementia (FTLD), Huntington’s disease (HD) and multiple sclerosis (MS); therefore, targeting its mechanisms might unveil new promising therapeutical strategies in their management [2][4][5].
While pharmacological treatments are available, there is an ongoing requirement for the development and discovery of new effective biomolecules that can enhance the quality of life for individuals afflicted by these diseases. Regarding this matter, a substantial body of research has been published on the advantageous neuroprotective effects of natural compounds that target this innate immune process [6][7]. Among all, phytocompounds within essential oils have demonstrated a promising capacity to counteract neuroinflammation and oxidative stress in preclinical models of neurotoxicity and neurodegenerative diseases, along with a significant improvement in cognitive processes such as learning and memory in these experimental systems [8][9].

2. Main Mediators of Neuroinflammation: The Role of Microglia and Astrocytes

Microglia and astrocytes are two key cellular regulators of inflammatory processes developing in the CNS. These two cell types can either exert pro-inflammatory or anti-inflammatory functions according to their polarization, classically categorized as M1 (pro-inflammatory) or M2 (anti- inflammatory) for microglia and A1 (pro-inflammatory) or A2 (anti-inflammatory) for astrocytes. Remarkably, many neuroprotective natural compounds function by rebalancing the pro-inflammatory phenotypes toward the anti-inflammatory ones [10][11]. It is important to underscore that categorizing these cells in this binary manner might not accurately represent the diverse phenotypes of microglia and astrocytes; thus, it is noteworthy to view them as existing along a spectrum rather than as entirely separate populations [2][12][13].

2.1. Microglia

Microglia, the macrophage-lineage cells of the CNS, represent the initial cell type that reacts to danger. These immune cells can shift from one phenotype to another in response to distinct environmental conditions within the CNS. When exposed to PAMPs or DAMPs, such as lipopolysaccharide (LPS) or reactive species, respectively, as well as IFN-γ, microglial cells are activated in the M1 phenotype, expressing pro-inflammatory signatures such as interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, chemokines, inducible nitric oxide synthase (iNOS), adenine dinucleotide phosphate (NADPH) oxidase and cyclooxygenase (COX)-2 [12][14]. This pro-inflammatory phenotype is associated with neural tissue damage [15]. NADPH oxidase produces ROS, such as superoxide anions, which in turn can combine with nitric oxide (NO) generated by iNOS to form peroxynitrite radicals [16]. When their concentration is greater than the cellular antioxidant capacity, reactive species are toxic, as they are capable of inducing DNA and protein damage as well as oxidizing the cellular membrane, resulting in lipid peroxidation and disruption of its properties, leading to necrotic cellular death [15][16][17]. TNF-α stimulates microglia in an autocrine manner, leading to an excessive release of glutamate, which consequently causes excitotoxic neuronal cell death [18]. Excessive glutamate dysregulates Ca2+ influx in neurons through hyper stimulation of the NMDA receptor, which in turn leads to RNS and ROS production, determining cellular death and exacerbating the neuroinflammatory response [19]. Several pathways are related to the pro-inflammatory switch of microglia.
LPS is recognized by, and thus activates, the Toll-like receptors (TLR)-4 signal pathway, which in turn culminates in a pro-inflammatory cascade and M1 microglia polarization. TLR4, activated by its ligand, leads to an enhanced activity of nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinases (MAPKs) JNK, ERK and p38, which enhance pro-inflammatory mediator transcription [7][12][20]. ROS and ion fluxes can trigger NOD-like receptor pyrin-domain-containing 3 (NLRP3)-inflammasome signaling and, hence, the neurotoxic M1 activation of microglia [21][22]. Of note, the NLRP3 inflammasome pathway is associated with the induction of a pro-inflammatory form of programmed cellular death called pyroptosis [23]. The Janus kinase/signal transducer and activator of transcription 1 (JAK1-2/STAT1) pathway is likewise implicated in M1 polarization. IFN-γ acts through this pathway, and when it binds to its receptor, it triggers STAT1 phosphorylation, increasing its transcriptional activity, which in turn upregulates pro- inflammatory genes [12][24][25]. Ultimately, contact-dependent Notch signaling drives microglia to the pro-inflammatory phenotype [26][27].
Given the pro-inflammatory role of the aforementioned signaling pathways, their pharmacological targeting may be beneficial in regulating the shift of microglia from the M1 to M2 polarization state. This modulation holds promise for the treatment of a range of brain diseases [28][29].
As already mentioned, induced by cytokines such as IL-4, IL-13 and IL-10, microglia can exhibit an anti-inflammatory phenotype. M2-polarized microglial cells release anti-inflammatory cytokines such as IL-10, Transforming Growth Factor (TGF)-β and IL-1R antagonists, which collectively act in opposition to their pro-inflammatory counterparts [30][31]. Additional factors expressed by M2 microglia associated with the resolution of neuroinflammation and neuroprotection include Arginase1 (Arg1), which suppresses NO production by competing with iNOS for arginine as a substrate and determines the production of molecules (i.e., polyamines) involved in tissue repair, cell proliferation and survival [30][32][33]; CD206 (also known as macrophage mannose receptor 1), a phagocytic receptor that binds myeloperoxidase and lysosomal hydrolases and, because of that, plays a pivotal role in the resolution of neuroinflammation [30][34]; and neurotrophic factors such as BDNF [30]. Several pathways are positively associated with M2 polarization. Some examples comprise JAK1/STAT6 (triggered by IL-4), the cannabinoid receptor 2 (CB2)/peroxisome proliferator-activated receptor gamma (PPAR-γ) axis [12][35][36], triggering receptor expressed on myeloid cells 2 (TREM2) signaling [20][37][38] and the PI3K/Akt cascade [39]. Notably, the role of this latter pathway on M1 to M2 polarization seems to be a function of specific Akt isoforms [40]. Boosting these pathways or, conversely, inhibiting the pro-inflammatory ones with exogenous compounds could be beneficial in the treatment of neuroinflammation associated with various brain disorders.

2.2. Astrocytes

Astrocytes play a fundamental role in maintaining brain homeostasis and are thus implicated in many CNS disorders. Notably, they contribute to blood–brain barrier (BBB) integrity, neuronal metabolism, synapse and neurotransmission regulation, potassium clearance, glymphatic flow control and host-defense mechanisms [41][42][43]. In the context of neuroinflammation, microglia and astrocytes interact to modulate the course of the response to an insult. Pro-inflammatory signals released by M1 microglia such as TNF-α, IL-1α and C1q stimulate reactive A1 astrocytes [44]. This particular astrocytal polarization exhibits a shared profile of secreted molecules to that of pro-inflammatory microglia, thus contributing to the augmentation of the neuroinflammatory burden [45][46].
Pointing out the remarkable role of microglia interaction with astrocytes in their A1 polarization, knock-out mice lacking microglia failed to induce A1 astrocytes after LPS treatment, while wild-type showed strong A1 induction [44]. Furthermore, it was demonstrated that the NLRP3 inflammasome pathway in microglia rather than in astrocytes is strongly associated with their pro-inflammatory shift and, accordingly, the knock-out of NLRP3 in microglia mitigates the neuronal dysfunction provoked by A1-like astrocytes, both in in vitro and in vivo settings [47][48]. A1 astrocytes release neurotoxic factors and promote glial scar formation, which overall can exert detrimental outcomes for brain repair and neuronal cell survival. For example, glial fibrillary acidic protein (GFAP) and chondroitin sulfate proteoglycans (CSPGs), the main components of glial scars, inhibit axonal regeneration [13][44][49][50]. Different signaling pathways and transcription factors are involved in glial scar formation driven by reactive astrocytes, including IL- 1/NF-κB, IL-6/STAT3, NOTCH/STAT3 and TGF-β/SMAD3; therefore, attenuating this neuroinflammatory-related process targeting these cell signaling cascades might be of benefit in CNS pathologies [51][52][53]. Analogously, M2 microglial cytokines trigger reactive A2 astrocytes, associated with anti-inflammatory cytokine release, neuroprotection and repair [44][45][46][54]. Pathways and transcription factors that are linked with enhanced A1 to A2 polarization include the PI3K/Akt axis [55] and STAT6, with this latter being associated with the upregulation of antioxidants genes in A2 astrocytes, such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Arg1 [56].
It is worth emphasizing that comprehending the mechanisms implicated in the regulation of neuroinflammation holds significant relevance in order to modulate this immune response through external molecules.

3. Neuroinflammation in Neurodegenerative Diseases: An Immunological Perspective

Neurodegeneration is a characteristic of numerous brain pathologies in which CNS functions deteriorate over time [5].
Neurodegenerative diseases place a significant burden on developed societies with aging populations. While research in this field is highly active, there remains a deficiency in comprehending the etiopathogenesis of these disorders, which is crucial for the discovery of new molecular targets and the development of therapies. In recent years, the role of neuroinflammation has emerged as a distinguishing feature of neurogenerative diseases, making it a new promising molecular process to focus on [5][57][58].
Notably during aging, a major risk factor for the development of neurodegenerative diseases, inflammatory processes rise in the brain (inflammaging) and there is a tendency for an increase in microglia M1/M2 ratio [33][59]. Additionally, the upregulation of gene signatures of M1-polarized microglia has been found in post-mortem tissues of patients with neurodegenerative disorders [60].
Along the same lines, an increase in A1 pro-inflammatory astrocytes has been identified in post-mortem neural tissues of individuals affected by AD, HT, PD, MS and ALS [44]. These findings suggest that neurodegenerative diseases develop in the context of a neuroinflammatory microenvironment.
Protein aggregates within neurons and in the extracellular space are a common characteristic and a hallmark of neurodegenerative diseases such as PD, AD, FTLD, HD and ALS [5][61]. Various studies clearly showed a link between protein aggregates in neurodegenerative diseases and neuroinflammation. Here, some examples of recent research and findings about this intricate association are provided.
Alpha-Synuclein (αSyn) aggregates (a pathological hallmark of PD) have been shown to lead to neuroinflammation and neurodegeneration through double-stranded DNA breaks and the induction of the DNA sensor GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) immune pathway, in a microglial and astrocyte mixed culture and a mouse model of PD [62]. Furthermore, in this study, the authors found STING upregulation in autopsied PD patients relative to healthy controls. These findings (associated with the amelioration of motor dysfunction symptoms in mice with STING knock-out) imply that this immune pathway, and therefore neuroinflammation, is an important feature in a neurodegenerative disorder such as PD [62][63]. Similarly, in an αSyn-driven mouse model of PD, the NLRP3 inflammasome cascade has been found to be upregulated in microglia [64]. The NLRP3 inflammasome pathway is also upregulated in microglia of mice models of PD driven by mitochondrial dysfunction and oxidative stress (thus independently of αSyn aggregates) and it precedes dopaminergic neuron degeneration and motor deficits [64]. Accordingly, NLRP3 pharmacological inhibition was found to be neuroprotective in these PD mice models. Furthermore, in PD patients, the upregulation of inflammasome markers has been evidenced in the substantia nigra of post-mortem brains [64]. Thus, taken together with other similar studies, these observations indicate that the aforementioned inflammatory signaling pathway is a key feature of this neurodegenerative disease [64][65]. Beta-amyloid (Aβ) aggregates, an AD pathological hallmark, induce the NLRP3 pathway through TLR4 in the BV-2 microglia cell line, and its conditioned medium reduces HT-22 neuronal cell line viability [66]. In another research study investigating the activation mechanism of the NLRP3 pathway by Aβ in primary microglia, the results revealed that Aβ aggregates initiate a process that involves the spleen tyrosine kinase (Syk)-mediated inactivation of AMPK. This inactivation leads to mitochondrial stress and an increase in the production of ROS, which subsequently triggers the NLRP3 cascade [67]. Interestingly, and in alignment with these findings, markers of pyroptosis (i.e., cleaved gasdermin D), an NLRP3-related inflammatory type of programmed cellular death, has been found to be upregulated in the post-mortem AD brain [68]. Moreover, the upregulation of genes and proteins associated with both inflammasome activation and pyroptosis is also observed in CNS post-mortem tissues of individuals with ALS and MS [69][70], underscoring the pivotal role of these processes as hallmarks in the landscape of neurodegenerative diseases. Of note, acetylcholinesterase (AChE) favors Aβ aggregate formation, thereby decreasing its activity associated with the inhibition of Aβ-fibrillogenesis [71].
Taking into account the central role of neuroinflammation in the abovementioned neurodegenerative diseases (described as examples of the various other brain disorders with neuroinflammation as a characteristic component), targeting immune pathway effectors or their biochemical activators (i.e., ROS, pathological protein aggregates, etc.) could represent a promising therapeutic strategy in order to prevent and ameliorate symptoms and the progression of neurodegenerative disorders.

References

  1. Kwon, H.S.; Koh, S.H. Neuroinflammation in Neurodegenerative Disorders: The Roles of Microglia and Astrocytes. Transl. Neurodegener. 2020, 9, 42.
  2. Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The Role and Consequences. Neurosci. Res. 2014, 79, 1–12.
  3. Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-Mediated Neuroinflammation in Neurodegenerative Diseases. Semin. Cell Dev. Biol. 2019, 94, 112–120.
  4. Palpagama, T.H.; Waldvogel, H.J.; Faull, R.L.M.; Kwakowsky, A. The Role of Microglia and Astrocytes in Huntington’s Disease. Front. Mol. Neurosci. 2019, 12, 258.
  5. Ransohoff, R.M. How Neuroinflammation Contributes to Neurodegeneration. Science 2016, 353, 777–783.
  6. Fanaro, G.B.; Marques, M.R.; Calaza, K.d.C.; Brito, R.; Pessoni, A.M.; Mendonça, H.R.; Lemos, D.E.d.A.; de Brito Alves, J.L.; de Souza, E.L.; Cavalcanti Neto, M.P. New Insights on Dietary Polyphenols for the Management of Oxidative Stress and Neuroinflammation in Diabetic Retinopathy. Antioxidants 2023, 12, 1237.
  7. Rahimifard, M.; Maqbool, F.; Moeini-Nodeh, S.; Niaz, K.; Abdollahi, M.; Braidy, N.; Nabavi, S.M.; Nabavi, S.F. Targeting the TLR4 Signaling Pathway by Polyphenols: A Novel Therapeutic Strategy for Neuroinflammation. Ageing Res. Rev. 2017, 36, 11–19.
  8. Capatina, L.; Napoli, E.M.; Ruberto, G.; Hritcu, L. Origanum vulgare ssp. Hirtum (Lamiaceae) Essential Oil Prevents Behavioral and Oxidative Stress Changes in the Scopolamine Zebrafish Model. Molecules 2021, 26, 7085.
  9. Postu, P.A.; Mihasan, M.; Gorgan, D.L.; Sadiki, F.Z.; El Idrissi, M.; Hritcu, L. Pinus Halepensis Essential Oil Ameliorates Aβ1-42-Induced Brain Injury by Diminishing Anxiety, Oxidative Stress, and Neuroinflammation in Rats. Biomedicines 2022, 10, 2300.
  10. Feng, J.; Wang, J.-X.; Du, Y.-H.; Liu, Y.; Zhang, W.; Chen, J.-F.; Liu, Y.-J.; Zheng, M.; Wang, K.-J.; He, G.-Q. Dihydromyricetin Inhibits Microglial Activation and Neuroinflammation by Suppressing NLRP3 Inflammasome Activation in APP/PS1 Transgenic Mice. CNS Neurosci. Ther. 2018, 24, 1207–1218.
  11. Liu, P.; Zhou, Y.; Shi, J.; Wang, F.; Yang, X.; Zheng, X.; Wang, Y.; He, Y.; Xie, X.; Pang, X. Myricetin Improves Pathological Changes in 3×Tg-AD Mice by Regulating the Mitochondria-NLRP3 Inflammasome-Microglia Channel by Targeting P38 MAPK Signaling Pathway. Phytomedicine 2023, 115, 154801.
  12. Guo, S.; Wang, H.; Yin, Y. Microglia Polarization from M1 to M2 in Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 815347.
  13. Liu, W.; Wang, Y.; Gong, F.; Rong, Y.; Luo, Y.; Tang, P.; Zhou, Z.; Zhou, Z.; Xu, T.; Jiang, T.; et al. Exosomes Derived from Bone Mesenchymal Stem Cells Repair Traumatic Spinal Cord Injury by Suppressing the Activation of A1 Neurotoxic Reactive Astrocytes. J. Neurotrauma 2019, 36, 469–484.
  14. Bordt, E.A.; Polster, B.M. NADPH Oxidase- and Mitochondria-Derived Reactive Oxygen Species in Proinflammatory Microglial Activation: A Bipartisan Affair? Free Radic. Biol. Med. 2014, 76, 34–46.
  15. Block, M.L.; Zecca, L.; Hong, J.-S. Microglia-Mediated Neurotoxicity: Uncovering the Molecular Mechanisms. Nat. Rev. Neurosci. 2007, 8, 57–69.
  16. Mosley, R.L.; Benner, E.J.; Kadiu, I.; Thomas, M.; Boska, M.D.; Hasan, K.; Laurie, C.; Gendelman, H.E. Neuroinflammation, Oxidative Stress and the Pathogenesis of Parkinson’s Disease. Clin. Neurosci. Res. 2006, 6, 261–281.
  17. Gaschler, M.M.; Stockwell, B.R. Lipid Peroxidation in Cell Death. Biochem. Biophys. Res. Commun. 2017, 482, 419–425.
  18. Takeuchi, H.; Jin, S.; Wang, J.; Zhang, G.; Kawanokuchi, J.; Kuno, R.; Sonobe, Y.; Mizuno, T.; Suzumura, A. Tumor Necrosis Factor-Alpha Induces Neurotoxicity via Glutamate Release from Hemichannels of Activated Microglia in an Autocrine Manner. J. Biol. Chem. 2006, 281, 21362–21368.
  19. Plotegher, N.; Filadi, R.; Pizzo, P.; Duchen, M.R. Excitotoxicity Revisited: Mitochondria on the Verge of a Nervous Breakdown. Trends Neurosci. 2021, 44, 342–351.
  20. Leri, M.; Vasarri, M.; Carnemolla, F.; Oriente, F.; Cabaro, S.; Stio, M.; Degl’Innocenti, D.; Stefani, M.; Bucciantini, M. EVOO Polyphenols Exert Anti-Inflammatory Effects on the Microglia Cell through TREM2 Signaling Pathway. Pharmaceuticals 2023, 16, 933.
  21. Blevins, H.M.; Xu, Y.; Biby, S.; Zhang, S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front. Aging Neurosci. 2022, 14, 879021.
  22. Chen, W.; Guo, C.; Huang, S.; Jia, Z.; Wang, J.; Zhong, J.; Ge, H.; Yuan, J.; Chen, T.; Liu, X.; et al. MitoQ Attenuates Brain Damage by Polarizing Microglia towards the M2 Phenotype through Inhibition of the NLRP3 Inflammasome after ICH. Pharmacol. Res. 2020, 161, 105122.
  23. Wang, S.; Yuan, Y.-H.; Chen, N.-H.; Wang, H.-B. The Mechanisms of NLRP3 Inflammasome/Pyroptosis Activation and Their Role in Parkinson’s Disease. Int. Immunopharmacol. 2019, 67, 458–464.
  24. Kann, O.; Almouhanna, F.; Chausse, B. Interferon γ: A Master Cytokine in Microglia-Mediated Neural Network Dysfunction and Neurodegeneration. Trends Neurosci. 2022, 45, 913–927.
  25. Rock, R.B.; Hu, S.; Deshpande, A.; Munir, S.; May, B.J.; Baker, C.A.; Peterson, P.K.; Kapur, V. Transcriptional Response of Human Microglial Cells to Interferon-Gamma. Genes. Immun. 2005, 6, 712–719.
  26. Yao, Y.-Y.; Li, R.; Guo, Y.-J.; Zhao, Y.; Guo, J.-Z.; Ai, Q.-L.; Zhong, L.-M.; Lu, D. Gastrodin Attenuates Lipopolysaccharide-Induced Inflammatory Response and Migration via the Notch-1 Signaling Pathway in Activated Microglia. Neuromol. Med. 2022, 24, 139–154.
  27. Zhang, Y.-H.; Wang, T.; Li, Y.-F.; Deng, Y.-N.; Shen, F.-G. Roles of the Notch Signaling Pathway and Microglia in Autism. Behav. Brain Res. 2023, 437, 114131.
  28. Capiralla, H.; Vingtdeux, V.; Zhao, H.; Sankowski, R.; Al-Abed, Y.; Davies, P.; Marambaud, P. Resveratrol Mitigates Lipopolysaccharide- and Aβ-Mediated Microglial Inflammation by Inhibiting the TLR4/NF-κB/STAT Signaling Cascade. J. Neurochem. 2012, 120, 461–472.
  29. Cui, W.; Sun, C.; Ma, Y.; Wang, S.; Wang, X.; Zhang, Y. Inhibition of TLR4 Induces M2 Microglial Polarization and Provides Neuroprotection via the NLRP3 Inflammasome in Alzheimer’s Disease. Front. Neurosci. 2020, 14, 444.
  30. Hu, X.; Leak, R.K.; Shi, Y.; Suenaga, J.; Gao, Y.; Zheng, P.; Chen, J. Microglial and Macrophage Polarization—New Prospects for Brain Repair. Nat. Rev. Neurol. 2015, 11, 56–64.
  31. Zhang, J.-M.; An, J. Cytokines, Inflammation, and Pain. Int. Anesthesiol. Clin. 2007, 45, 27–37.
  32. Li, Z.; Wang, L.; Ren, Y.; Huang, Y.; Liu, W.; Lv, Z.; Qian, L.; Yu, Y.; Xiong, Y. Arginase: Shedding Light on the Mechanisms and Opportunities in Cardiovascular Diseases. Cell Death Discov. 2022, 8, 413.
  33. Tang, Y.; Li, T.; Li, J.; Yang, J.; Liu, H.; Zhang, X.J.; Le, W. Jmjd3 Is Essential for the Epigenetic Modulation of Microglia Phenotypes in the Immune Pathogenesis of Parkinson’s Disease. Cell Death Differ. 2014, 21, 369–380.
  34. Gazi, U.; Martinez-Pomares, L. Influence of the Mannose Receptor in Host Immune Responses. Immunobiology 2009, 214, 554–561.
  35. Rahimian, R.; Belliveau, C.; Chen, R.; Mechawar, N. Microglial Inflammatory-Metabolic Pathways and Their Potential Therapeutic Implication in Major Depressive Disorder. Front. Psychiatry 2022, 13, 871997.
  36. He, Y.; Gao, Y.; Zhang, Q.; Zhou, G.; Cao, F.; Yao, S. IL-4 Switches Microglia/Macrophage M1/M2 Polarization and Alleviates Neurological Damage by Modulating the JAK1/STAT6 Pathway Following ICH. Neuroscience 2020, 437, 161–171.
  37. Wang, Y.; Lin, Y.; Wang, L.; Zhan, H.; Luo, X.; Zeng, Y.; Wu, W.; Zhang, X.; Wang, F. TREM2 Ameliorates Neuroinflammatory Response and Cognitive Impairment via PI3K/AKT/FoxO3a Signaling Pathway in Alzheimer’s Disease Mice. Aging 2020, 12, 20862–20879.
  38. Zhang, J.; Zheng, Y.; Luo, Y.; Du, Y.; Zhang, X.; Fu, J. Curcumin Inhibits LPS-Induced Neuroinflammation by Promoting Microglial M2 Polarization via TREM2/ TLR4/ NF-κB Pathways in BV2 Cells. Mol. Immunol. 2019, 116, 29–37.
  39. Liu, W.; Rong, Y.; Wang, J.; Zhou, Z.; Ge, X.; Ji, C.; Jiang, D.; Gong, F.; Li, L.; Chen, J.; et al. Exosome-Shuttled miR-216a-5p from Hypoxic Preconditioned Mesenchymal Stem Cells Repair Traumatic Spinal Cord Injury by Shifting Microglial M1/M2 Polarization. J. Neuroinflamm. 2020, 17, 47.
  40. Vergadi, E.; Ieronymaki, E.; Lyroni, K.; Vaporidi, K.; Tsatsanis, C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J. Immunol. 2017, 198, 1006–1014.
  41. Bellot-Saez, A.; Stevenson, R.; Kékesi, O.; Samokhina, E.; Ben-Abu, Y.; Morley, J.W.; Buskila, Y. Neuromodulation of Astrocytic K+ Clearance. Int. J. Mol. Sci. 2021, 22, 2520.
  42. Gomolka, R.S.; Hablitz, L.M.; Mestre, H.; Giannetto, M.; Du, T.; Hauglund, N.L.; Xie, L.; Peng, W.; Martinez, P.M.; Nedergaard, M.; et al. Loss of Aquaporin-4 Results in Glymphatic System Dysfunction via Brain-Wide Interstitial Fluid Stagnation. eLife 2023, 12, e82232.
  43. Phatnani, H.; Maniatis, T. Astrocytes in Neurodegenerative Disease. Cold Spring Harb. Perspect. Biol. 2015, 7, a020628.
  44. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.-S.; Peterson, T.C.; et al. Neurotoxic Reactive Astrocytes Are Induced by Activated Microglia. Nature 2017, 541, 481–487.
  45. Liu, L.-R.; Liu, J.-C.; Bao, J.-S.; Bai, Q.-Q.; Wang, G.-Q. Interaction of Microglia and Astrocytes in the Neurovascular Unit. Front. Immunol. 2020, 11, 1024.
  46. Xie, L.; Zhang, N.; Zhang, Q.; Li, C.; Sandhu, A.F.; Iii, G.W.; Lin, S.; Lv, P.; Liu, Y.; Wu, Q.; et al. Inflammatory Factors and Amyloid β-Induced Microglial Polarization Promote Inflammatory Crosstalk with Astrocytes. Aging 2020, 12, 22538–22549.
  47. Li, S.; Fang, Y.; Zhang, Y.; Song, M.; Zhang, X.; Ding, X.; Yao, H.; Chen, M.; Sun, Y.; Ding, J.; et al. Microglial NLRP3 Inflammasome Activates Neurotoxic Astrocytes in Depression-like Mice. Cell Rep. 2022, 41, 111532.
  48. Xiao, T.; Ji, H.; Shangguan, X.; Qu, S.; Cui, Y.; Xu, J. NLRP3 Inflammasome of Microglia Promotes A1 Astrocyte Transformation, Neo-Neuron Decline and Cognition Impairment in Endotoxemia. Biochem. Biophys. Res. Commun. 2022, 602, 1–7.
  49. Sofroniew, M.V. Molecular Dissection of Reactive Astrogliosis and Glial Scar Formation. Trends Neurosci. 2009, 32, 638–647.
  50. Wang, H.; Song, G.; Chuang, H.; Chiu, C.; Abdelmaksoud, A.; Ye, Y.; Zhao, L. Portrait of Glial Scar in Neurological Diseases. Int. J. Immunopathol. Pharmacol. 2018, 31, 2058738418801406.
  51. Hamby, M.E.; Sofroniew, M.V. Reactive Astrocytes as Therapeutic Targets for CNS Disorders. Neurotherapeutics 2010, 7, 494–506.
  52. Qian, D.; Li, L.; Rong, Y.; Liu, W.; Wang, Q.; Zhou, Z.; Gu, C.; Huang, Y.; Zhao, X.; Chen, J.; et al. Blocking Notch Signal Pathway Suppresses the Activation of Neurotoxic A1 Astrocytes after Spinal Cord Injury. Cell Cycle 2019, 18, 3010–3029.
  53. Wu, M.; Wang, L.; Li, F.; Hu, R.; Ma, J.; Zhang, K.; Cheng, X. Resveratrol Downregulates STAT3 Expression and Astrocyte Activation in Primary Astrocyte Cultures of Rat. Neurochem. Res. 2020, 45, 455–464.
  54. Chang, J.; Qian, Z.; Wang, B.; Cao, J.; Zhang, S.; Jiang, F.; Kong, R.; Yu, X.; Cao, X.; Yang, L.; et al. Transplantation of A2 Type Astrocytes Promotes Neural Repair and Remyelination after Spinal Cord Injury. Cell Commun. Signal 2023, 21, 37.
  55. Li, T.; Liu, T.; Chen, X.; Li, L.; Feng, M.; Zhang, Y.; Wan, L.; Zhang, C.; Yao, W. Microglia Induce the Transformation of A1/A2 Reactive Astrocytes via the CXCR7/PI3K/Akt Pathway in Chronic Post-Surgical Pain. J. Neuroinflamm. 2020, 17, 211.
  56. Neal, M.; Luo, J.; Harischandra, D.S.; Gordon, R.; Sarkar, S.; Jin, H.; Anantharam, V.; Désaubry, L.; Kanthasamy, A.; Kanthasamy, A. Prokineticin-2 Promotes Chemotaxis and Alternative A2 Reactivity of Astrocytes. Glia 2018, 66, 2137–2157.
  57. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS Neurodegenerative Diseases. Immunology 2018, 154, 204–219.
  58. Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W.; Li, J. Aging and Aging-Related Diseases: From Molecular Mechanisms to Interventions and Treatments. Signal Transduct. Target. Ther. 2022, 7, 391.
  59. Scheiblich, H.; Trombly, M.; Ramirez, A.; Heneka, M.T. Neuroimmune Connections in Aging and Neurodegenerative Diseases. Trends Immunol. 2020, 41, 300–312.
  60. Li, M.D.; Burns, T.C.; Morgan, A.A.; Khatri, P. Integrated Multi-Cohort Transcriptional Meta-Analysis of Neurodegenerative Diseases. Acta Neuropathol. Commun. 2014, 2, 93.
  61. Ross, C.A.; Poirier, M.A. Protein Aggregation and Neurodegenerative Disease. Nat. Med. 2004, 10 (Suppl. S7), S10–S17.
  62. Hinkle, J.T.; Patel, J.; Panicker, N.; Karuppagounder, S.S.; Biswas, D.; Belingon, B.; Chen, R.; Brahmachari, S.; Pletnikova, O.; Troncoso, J.C.; et al. STING Mediates Neurodegeneration and Neuroinflammation in Nigrostriatal α-Synucleinopathy. Proc. Natl. Acad. Sci. USA 2022, 119, e2118819119.
  63. Gulen, M.F.; Samson, N.; Keller, A.; Schwabenland, M.; Liu, C.; Glück, S.; Thacker, V.V.; Favre, L.; Mangeat, B.; Kroese, L.J.; et al. cGAS-STING Drives Ageing-Related Inflammation and Neurodegeneration. Nature 2023, 620, 374–380.
  64. Gordon, R.; Albornoz, E.A.; Christie, D.C.; Langley, M.R.; Kumar, V.; Mantovani, S.; Robertson, A.A.B.; Butler, M.S.; Rowe, D.B.; O’Neill, L.A.; et al. Inflammasome Inhibition Prevents α-Synuclein Pathology and Dopaminergic Neurodegeneration in Mice. Sci. Transl. Med. 2018, 10, eaah4066.
  65. Lee, E.; Hwang, I.; Park, S.; Hong, S.; Hwang, B.; Cho, Y.; Son, J.; Yu, J.-W. MPTP-Driven NLRP3 Inflammasome Activation in Microglia Plays a Central Role in Dopaminergic Neurodegeneration. Cell Death Differ. 2019, 26, 213–228.
  66. Liu, Y.; Dai, Y.; Li, Q.; Chen, C.; Chen, H.; Song, Y.; Hua, F.; Zhang, Z. Beta-Amyloid Activates NLRP3 Inflammasome via TLR4 in Mouse Microglia. Neurosci. Lett. 2020, 736, 135279.
  67. Jung, E.S.; Suh, K.; Han, J.; Kim, H.; Kang, H.-S.; Choi, W.-S.; Mook-Jung, I. Amyloid-β Activates NLRP3 Inflammasomes by Affecting Microglial Immunometabolism through the Syk-AMPK Pathway. Aging Cell 2022, 21, e13623.
  68. Moonen, S.; Koper, M.J.; Van Schoor, E.; Schaeverbeke, J.M.; Vandenberghe, R.; von Arnim, C.A.F.; Tousseyn, T.; De Strooper, B.; Thal, D.R. Pyroptosis in Alzheimer’s Disease: Cell Type-Specific Activation in Microglia, Astrocytes and Neurons. Acta Neuropathol. 2023, 145, 175–195.
  69. McKenzie, B.A.; Mamik, M.K.; Saito, L.B.; Boghozian, R.; Monaco, M.C.; Major, E.O.; Lu, J.-Q.; Branton, W.G.; Power, C. Caspase-1 Inhibition Prevents Glial Inflammasome Activation and Pyroptosis in Models of Multiple Sclerosis. Proc. Natl. Acad. Sci. USA 2018, 115, E6065–E6074.
  70. Van Schoor, E.; Ospitalieri, S.; Moonen, S.; Tomé, S.O.; Ronisz, A.; Ok, O.; Weishaupt, J.; Ludolph, A.C.; Van Damme, P.; Van Den Bosch, L.; et al. Increased Pyroptosis Activation in White Matter Microglia Is Associated with Neuronal Loss in ALS Motor Cortex. Acta Neuropathol. 2022, 144, 393–411.
  71. Bartolini, M.; Bertucci, C.; Cavrini, V.; Andrisano, V. Beta-Amyloid Aggregation Induced by Human Acetylcholinesterase: Inhibition Studies. Biochem. Pharmacol. 2003, 65, 407–416.
More
Information
Subjects: Neurosciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 180
Revisions: 2 times (View History)
Update Date: 08 Feb 2024
1000/1000
Video Production Service