Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer’s Disease
Abstract
:1. Introduction
2. Mechanisms of Alzheimer’s Disease Pathogenesis
2.1. Role of Oxidative Stress
2.2. Role of Neuroinflammation
2.3. Contribution of Glymphatic System
2.4. Role of Cerebral Blood Flow
3. Discussion
Author Contributions
Funding
Conflicts of Interest
References
- Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef] [PubMed]
- Harper, L.C. 2022 Alzheimer’s disease facts and figures. Alzheimers Dement. 2022, 18, 700–789. [Google Scholar] [CrossRef]
- Mendez, M.F. Early-Onset Alzheimer Disease. Neurol. Clin. 2017, 35, 263–281. [Google Scholar] [CrossRef] [PubMed]
- Soria Lopez, J.A.; González, H.M.; Léger, G.C. Alzheimer’s disease. Handb. Clin. Neurol. 2019, 167, 231–255. [Google Scholar] [CrossRef] [PubMed]
- Porsteinsson, A.P.; Isaacson, R.S.; Knox, S.; Sabbagh, M.N.; Rubino, I. Diagnosis of Early Alzheimer’s Disease: Clinical Practice in 2021. J. Prev. Alzheimers Dis. 2021, 8, 371–386. [Google Scholar] [CrossRef]
- Kazim, S.F.; Iqbal, K. Neurotrophic factor small-molecule mimetics mediated neuroregeneration and synaptic repair: Emerging therapeutic modality for Alzheimer’s disease. Mol. Neurodegener. 2016, 11, 50. [Google Scholar] [CrossRef]
- Tiwari, S.; Atluri, V.; Kaushik, A.; Yndart, A.; Nair, M. Alzheimer’s disease: Pathogenesis, diagnostics, and therapeutics. Int. J. Nanomed. 2019, 14, 5541–5554. [Google Scholar] [CrossRef]
- Hampel, H.; Hardy, J.; Blennow, K.; Chen, C.; Perry, G.; Kim, S.H.; Villemagne, V.L.; Aisen, P.; Vendruscolo, M.; Iwatsubo, T.; et al. The Amyloid-β Pathway in Alzheimer’s Disease. Mol. Psychiatry 2021, 26, 5481–5503. [Google Scholar] [CrossRef]
- Khan, S.; Barve, K.H.; Kumar, M.S. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer’s Disease. Curr. Neuropharmacol. 2020, 18, 1106–1125. [Google Scholar] [CrossRef]
- Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chételat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer disease. Nat. Rev. Dis. Primers 2021, 7, 33. [Google Scholar] [CrossRef]
- Dubois, B.; Feldman, H.H.; Jacova, C.; Hampel, H.; Molinuevo, J.L.; Blennow, K.; DeKosky, S.T.; Gauthier, S.; Selkoe, D.; Bateman, R.; et al. Advancing research diagnostic criteria for Alzheimer’s disease: The IWG-2 criteria. Lancet Neurol. 2014, 13, 614–629. [Google Scholar] [CrossRef]
- Hampel, H.; Cummings, J.; Blennow, K.; Gao, P.; Jack, C.R., Jr.; Vergallo, A. Developing the ATX(N) classification for use across the Alzheimer disease continuum. Nat. Rev. Neurol. 2021, 17, 580–589. [Google Scholar] [CrossRef]
- Meziane, H.; Dodart, J.C.; Mathis, C.; Little, S.; Clemens, J.; Paul, S.M.; Ungerer, A. Memory-enhancing effects of secreted forms of the β-amyloid precursor protein in normal and amnestic mice. Proc. Natl. Acad. Sci. USA 1998, 95, 2683–12688. [Google Scholar] [CrossRef]
- Kimberly, W.T.; Zheng, J.B.; Guénette, S.Y.; Selkoe, D.J. The intracellular domain of the β-amyloid precursor protein is stabilized by Fe65 and translocates to the nucleus in a notch-like manner. J. Biol. Chem. 2001, 276, 40288–40292. [Google Scholar] [CrossRef]
- Bekris, L.M.; Yu, C.E.; Bird, T.D.; Tsuang, D.W. Genetics of Alzheimer Disease. J. Geriatr. Psychiatry Neurol. 2010, 23, 213–227. [Google Scholar] [CrossRef]
- Sun, Q.; Xie, N.; Tang, B.; Li, R.; Shen, Y. Alzheimer’s Disease: From Genetic Variants to the Distinct Pathological Mechanisms. Front. Mol. Neurosci. 2017, 10, 2017. [Google Scholar] [CrossRef]
- Hoogmartens, J.; Cacace, R.; Broeckhoven, C.V. Insight into the genetic etiology of Alzheimer’s disease: A comprehensive review of the role of rare variants. Alzheimers Dement. Diagn. Assess. Dis. Monit. 2021, 13, e12155. [Google Scholar] [CrossRef]
- Tu, S.; Okamoto, S.; Lipton, S.A.; Xu, H. Oligomeric Aβ-induced synaptic dysfunction in Alzheimer’s disease. Mol. Neurodegener. 2014, 9, 48. [Google Scholar] [CrossRef]
- Grimm, M.O.W.; Hartmann, T. Recent understanding of the molecular mechanisms of Alzheimer’s disease. J. Addict. Res. Ther. 2012, 5, 1–27. [Google Scholar] [CrossRef]
- Pooler, A.M.; Noble, W.; Hanger, D.P. A role for tau at the synapse in Alzheimer’s disease pathogenesis. Neuropharmacology 2014, 76 Pt A, 1–8. [Google Scholar] [CrossRef]
- Eftekharzadeh, B.; Daigle, J.G.; Kapinos, L.E.; Coyne, A.; Schiantarelli, J.; Carlomagno, Y.; Cook, C.; Miller, S.J.; Dujardin, S.; Amaral, A.S.; et al. Tau Protein Disrupts Nucleocytoplasmic Transport in Alzheimer’s Disease. Neuron 2018, 99, 925–940.e7. [Google Scholar] [CrossRef] [PubMed]
- Kent, S.A.; Spires-Jones, T.L.; Durrant, C.S. The physiological roles of tau and Aβ: Implications for Alzheimer’s disease pathology and therapeutics. Acta Neuropathol. 2020, 140, 417–447. [Google Scholar] [CrossRef] [PubMed]
- Lee, V.M.; Goedert, M.; Trojanowski, J.Q. Neurodegenerative tauopathies. Annu. Rev. Neurosci. 2001, 24, 1121–1159. [Google Scholar] [CrossRef] [PubMed]
- de Strooper, B.; Woodgett, J. Alzheimer’s disease: Mental plaque removal. Nature 2003, 423, 392–393. [Google Scholar] [CrossRef]
- Kametani, F.; Hasegawa, M. Reconsideration of Amyloid Hypothesis and Tau Hypothesis in Alzheimer’s Disease. Front. Neurosci. 2018, 12, 25. [Google Scholar] [CrossRef]
- Ostrowitzki, S.; Deptula, D.; Thurfjell, L.; Barkhof, F.; Bohrmann, B.; Brooks, D.J.; Klunk, W.E.; Ashford, E.; Yoo, K.; Xu, Z.X.; et al. Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch. Neurol. 2012, 69, 198–207. [Google Scholar] [CrossRef]
- Giacobini, E.; Gold, G. Alzheimer disease therapy--moving from amyloid-β to tau. Nat. Rev. Neurol. 2013, 9, 677–686. [Google Scholar] [CrossRef]
- Doody, R.S.; Thomas, R.G.; Farlow, M.; Iwatsubo, T.; Vellas, B.; Joffe, S.; Kieburtz, K.; Raman, R.; Sun, X.; Aisen, P.S.; et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 311–321. [Google Scholar] [CrossRef]
- Salloway, S.; Sperling, R.; Fox, N.C.; Blennow, K.; Klunk, W.; Raskind, M.; Sabbagh, M.; Honig, L.S.; Porsteinsson, A.P.; Ferris, S.; et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 322–333. [Google Scholar] [CrossRef]
- Kim, J.; Onstead, L.; Randle, S.; Price, R.; Smithson, L.; Zwizinski, C.; Dickson, D.W.; Golde, T.; McGowan, E. Aβ40 inhibits amyloid deposition in vivo. J. Neurosci. 2007, 27, 627–633. [Google Scholar] [CrossRef]
- Kim, J.; Chakrabarty, P.; Hanna, A.; March, A.; Dickson, D.W.; Borchelt, D.R.; Golde, T.; Janus, C. Normal cognition in transgenic BRI2-Aβ mice. Mol. Neurodegener. 2013, 8, 15. [Google Scholar] [CrossRef]
- Price, J.L.; McKeel, D.W., Jr.; Buckles, V.D.; Roe, C.M.; Xiong, C.; Grundman, M.; Hansen, L.A.; Petersen, R.C.; Parisi, J.E.; Dickson, D.W.; et al. Neuropathology of nondemented aging: Presumptive evidence for preclinical Alzheimer disease. Neurobiol. Aging 2009, 30, 1026–1036. [Google Scholar] [CrossRef]
- Chételat, G.; La Joie, R.; Villain, N.; Perrotin, A.; de La Sayette, V.; Eustache, F.; Vandenberghe, R. Amyloid imaging in cognitively normal individuals, at-risk populations and preclinical Alzheimer’s disease. Neuroimage Clin. 2013, 2, 356–365. [Google Scholar] [CrossRef]
- Bejanin, A.; Schonhaut, D.R.; La Joie, R.; Kramer, J.H.; Baker, S.L.; Sosa, N.; Ayakta, N.; Cantwell, A.; Janabi, M.; Lauriola, M.; et al. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 2017, 140, 3286–3300. [Google Scholar] [CrossRef]
- Hochgräfe, K.; Sydow, A.; Mandelkow, E.M. Regulatable transgenic mouse models of Alzheimer disease: Onset, reversibility and spreading of Tau pathology. FEBS J. 2013, 280, 4371–4381. [Google Scholar] [CrossRef]
- Rusu, P.; Jansen, A.; Soba, P.; Kirsch, J.; Löwer, A.; Merdes, G.; Kuan, Y.; Jung, A.; Beyreuther, K.; Kjaerulff, O.; et al. Axonal accumulation of synaptic markers in APP transgenic Drosophila depends on the NPTY motif and is paralleled by defects in synaptic plasticity. Eur. J. Neurosci. 2007, 25, 1079–1086. [Google Scholar] [CrossRef]
- Tini, G.; Scagliola, R.; Monacelli, F.; La Malfa, G.; Porto, I.; Brunelli, C.; Rosa, G.M. Alzheimer’s Disease and Cardiovascular Disease: A Particular Association. Cardiol. Res. Pract. 2020, 2020, 2617970. [Google Scholar] [CrossRef]
- Peng, L.; Bestard-Lorigados, I.; Song, W. The synapse as a treatment avenue for Alzheimer’s Disease. Mol. Psychiatry 2022, 27, 2940–2949. [Google Scholar] [CrossRef]
- Koffie, R.M.; Meyer-Luehmann, M.; Hashimoto, T.; Adams, K.W.; Mielke, M.L.; Garcia-Alloza, M.; Micheva, K.D.; Smith, S.J.; Kim, M.L.; Lee, V.M.; et al. Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc. Natl. Acad. Sci. USA 2009, 106, 4012–4017. [Google Scholar] [CrossRef]
- Kopeikina, K.J.; Polydoro, M.; Tai, H.C.; Yaeger, E.; Carlson, G.A.; Pitstick, R.; Hyman, B.T.; Spires-Jones, T.L. Synaptic alterations in the rTg4510 mouse model of tauopathy. J. Comp. Neurol. 2013, 521, 1334–1353. [Google Scholar] [CrossRef]
- Mucke, L.; Masliah, E.; Yu, G.Q.; Mallory, M.; Rockenstein, E.M.; Tatsuno, G.; Hu, K.; Kholodenko, D.; Johnson-Wood, K.; McConlogue, L. High-level neuronal expression of Aβ1–42 in wild-type human amyloid protein precursor transgenic mice: Synaptotoxicity without plaque formation. J. Neurosci. 2000, 20, 4050–4058. [Google Scholar] [CrossRef] [PubMed]
- Angioni, D.; Delrieu, J.; Hansson, O.; Fillit, H.; Aisen, P.; Cummings, J.; Sims, J.R.; Braunstein, J.B.; Sabbagh, M.; Bittner, T.; et al. Blood Biomarkers from Research Use to Clinical Practice: What Must Be Done? A Report from the EU/US CTAD Task Force. J. Prev. Alzheimers Dis. 2022, 9, 569–579. [Google Scholar] [CrossRef] [PubMed]
- Klyucherev, T.O.; Olszewski, P.; Shalimova, A.A.; Chubarev, V.N.; Tarasov, V.V.; Attwood, M.M.; Syvanen, S.; Schioth, H.B. Advances in the development of new biomarkers for Alzheimer’s disease. Transl. Neurodegener. 2022, 25, 2022. [Google Scholar] [CrossRef] [PubMed]
- Salehpour, F.; Mahmoudi, J.; Kamari, F.; Sadigh-Eteghad, S.; Rasta, S.H.; Hamblin, M.R. Brain Photobiomodulation Therapy: A Narrative Review. Mol. Neurobiol. 2018, 55, 6601–6636. [Google Scholar] [CrossRef]
- Hennessy, M.; Hamblin, M.R. Photobiomodulation and the brain: A new paradigm. J. Opt. 2017, 19, 013003. [Google Scholar] [CrossRef]
- Lapchak, P.A.; Wei, J.; Zivin, J.A. Transcranial infrared laser therapy improves clinical rating scores after embolic strokes in rabbits. Stroke 2004, 35, 1985–1988. [Google Scholar] [CrossRef]
- Karu, T.I. Molecular mechanism of the therapeutic effect of low-intensity laser radiation. Lasers Life Sci. 1988, 2, 53–74. [Google Scholar]
- Wang, Y.; Huang, Y.Y.; Wang, Y.; Lyu, P.; Hamblin, M.R. Photobiomodulation of human adipose-derived stem cells using 810 nm and 980 nm lasers operates via different mechanisms of action. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 441–449. [Google Scholar] [CrossRef]
- Martins, I.J.; Hone, E.; Foster, J.K.; Sünram-Lea, S.I.; Gnjec, A.; Fuller, S.J.; Nolan, D.; Gandy, S.E.; Martins, R.N. Apolipoprotein E, cholesterol metabolism, diabetes, and the convergence of risk factors for Alzheimer’s disease and cardiovascular disease. Mol. Psychiatry 2006, 11, 721–736. [Google Scholar] [CrossRef]
- Naganawa, S.; Taoka, T. The Glymphatic System: A Review of the Challenges in Visualizing its Structure and Function with MR Imaging. Magn. Reson. Med. Sci. 2022, 21, 182–194. [Google Scholar] [CrossRef]
- Jin, B.J.; Smith, A.J.; Verkman, A.S. Spatial model of convective solute transport in brain extracellular space does not support a “glymphatic” mechanism. J. Gen. Physiol. 2016, 148, 489–501. [Google Scholar] [CrossRef]
- Hamblin, M.R. The role of nitric oxide in low level light therapy. In Mechanisms for Low-Light Therapy III; SPIE: Bellingham, WA, USA, 2008; Volume 6846, p. 684602. [Google Scholar] [CrossRef]
- Gao, X.; Xing, D. Molecular mechanisms of cell proliferation induced by low power laser irradiation. J. Biomed. Sci. 2009, 16, 4. [Google Scholar] [CrossRef]
- de Freitas, L.F.; Hamblin, M.R. Proposed Mechanisms of Photobiomodulation or Low-Level Light Therapy. IEEE J. Sel. Top. Quantum Electron. 2016, 22, 7000417. [Google Scholar] [CrossRef]
- Nunnari, J.; Suomalainen, A. Mitochondria: In sickness and in health. Cell 2012, 148, 1145–1159. [Google Scholar] [CrossRef]
- Schwarz, T.L. Mitochondrial trafficking in neurons. Cold Spring Harb. Perspect. Biol. 2013, 5, a011304. [Google Scholar] [CrossRef]
- Chen, A.C.; Arany, P.R.; Huang, Y.Y.; Tomkinson, E.M.; Sharma, S.K.; Kharkwal, G.B.; Saleem, T.; Mooney, D.; Yull, F.E.; Blackwell, T.S.; et al. Low-level laser therapy activates NF-kB via generation of reactive oxygen species in mouse embryonic fibroblasts. PLoS ONE 2011, 6, e22453. [Google Scholar] [CrossRef]
- Tafur, J.; Mills, P.J. Low-intensity light therapy: Exploring the role of redox mechanisms. Photomed. Laser Surg. 2008, 26, 323–328. [Google Scholar] [CrossRef]
- Chen, A.C.; Huang, Y.Y.; Sharma, S.K.; Hamblin, M.R. Effects of 810-nm laser on murine bone-marrow-derived dendritic cells. Photomed. Laser Surg. 2011, 29, 383–389. [Google Scholar] [CrossRef]
- Hamblin, M.R. Mechanisms and applications of the anti-inflammatory effects of photobiomodulation. AIMS Biophys. 2017, 4, 337–361. [Google Scholar] [CrossRef]
- Litscher, G.; Min, L.; Passegger, C.A.; Litscher, D.; Li, M.; Wang, M.; Ghaffari-Tabrizi-Wizsy, N.; Stelzer, I.; Feigl, G.; Gaischek, I.; et al. Transcranial yellow, red, and infrared laser and LED stimulation: Changes of vascular parameters in a chick embryo model. Integr. Med. Int. 2015, 2, 80–89. [Google Scholar] [CrossRef]
- Shefer, G.; Partridge, T.A.; Heslop, L.; Gross, J.G.; Oron, U.; Halevy, O. Low-energy laser irradiation promotes the survival and cell cycle entry of skeletal muscle satellite cells. J. Cell Sci. 2002, 115, 1461–1469. [Google Scholar] [CrossRef]
- Liang, H.L.; Whelan, H.T.; Eells, J.T.; Wong-Riley, M.T. Near-infrared light via light-emitting diode treatment is therapeutic against rotenone- and 1-methyl-4-phenylpyridinium ion-induced neurotoxicity. Neuroscience 2008, 153, 963–974. [Google Scholar] [CrossRef] [PubMed]
- Telerman, A.; Lapter, S.; Sharabi, A.; Zinger, H.; Mozes, E. Induction of hippocampal neurogenesis by a tolerogenic peptide that ameliorates lupus manifestations. J. Neuroimmunol. 2011, 232, 151–157. [Google Scholar] [CrossRef] [PubMed]
- Nizamutdinov, D.; Qi, X.; Berman, M.H.; Dougal, G.; Dayawansa, S.; Wu, E.; Yi, S.S.; Stevens, A.B.; Huang, J.H. Transcranial Near Infrared Light Stimulations Improve Cognition in Patients with Dementia. Aging Dis. 2021, 12, 954–963. [Google Scholar] [CrossRef] [PubMed]
- Valverde, A.; Mitrofanis, J. Photobiomodulation for Hypertension and Alzheimer’s Disease. J. Alzheimers Dis 2022, 90, 1045–1055. [Google Scholar] [CrossRef]
- Iadecola, C.; Davisson, R.L. Hypertension and cerebrovascular dysfunction. Cell Metab. 2008, 7, 476–484. [Google Scholar] [CrossRef]
- de Jong, D.L.K.; de Heus, R.A.A.; Rijpma, A.; Donders, R.; Olde Rikkert, M.G.M.; Günther, M.; Lawlor, B.A.; van Osch, M.J.P.; Claassen, J.A.H.R. Effects of Nilvadipine on Cerebral Blood Flow in Patients with Alzheimer Disease. Hypertension 2019, 74, 413–420. [Google Scholar] [CrossRef]
- Gupta, A.; Iadecola, C. Impaired Aβ clearance: A potential link between atherosclerosis and Alzheimer’s disease. Front. Aging Neurosci. 2015, 7, 115. [Google Scholar] [CrossRef]
- Carrano, A.; Hoozemans, J.J.; van der Vies, S.M.; Rozemuller, A.J.; van Horssen, J.; de Vries, H.E. Amyloid Beta induces oxidative stress-mediated blood-brain barrier changes in capillary amyloid angiopathy. Antioxid. Redox Signal. 2011, 15, 1167–1178. [Google Scholar] [CrossRef]
- Ando, T.; Xuan, W.; Xu, T.; Dai, T.; Sharma, S.K.; Kharkwal, G.B.; Huang, Y.Y.; Wu, Q.; Whalen, W.J.; Sato, S.; et al. Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice. PLoS ONE 2011, 6, e26212. [Google Scholar] [CrossRef]
- Wang, X.; Li, X.; Zuo, X.; Liang, Z.; Ding, T.; Li, K.; Ma, Y.; Li, P.; Zhu, Z.; Ju, C.; et al. Photobiomodulation inhibits the activation of neurotoxic microglia and astrocytes by inhibiting Lcn2/JAK2-STAT3 crosstalk after spinal cord injury in male rats. J. Neuroinflamm. 2021, 18, 256. [Google Scholar] [CrossRef]
- Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
- Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
- Kumar, S.; Pandey, A.K. Free radicals: Health implications and their mitigation by herbals. Br. J. Med. Med. Res. 2015, 7, 438–457. [Google Scholar] [CrossRef]
- Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef]
- Young, I.S.; Woodside, J.V. Antioxidants in health and disease. J. Clin. Pathol. 2001, 54, 176–186. [Google Scholar] [CrossRef]
- Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef]
- Bonda, D.J.; Wang, X.; Lee, H.G.; Smith, M.A.; Perry, G.; Zhu, X. Neuronal failure in Alzheimer’s disease: A view through the oxidative stress looking-glass. Neurosci. Bull. 2014, 30, 243–252. [Google Scholar] [CrossRef]
- Grimm, A.; Eckert, A. Brain aging and neurodegeneration: From a mitochondrial point of view. J. Neurochem. 2017, 143, 418–431. [Google Scholar] [CrossRef]
- Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef]
- Reybier, K.; Ayala, S.; Alies, B.; Rodrigues, J.V.; Rodriguez, S.B.; la Penna, G.; Collin, F.; Gomes, C.M.; Hureau, C.; Faller, P. Free Superoxide is an Intermediate in the Production of H2O2 by Copper(I)-Aβ Peptide and O2. Angew. Chem. Int. Ed. Engl. 2016, 55, 1085–1089. [Google Scholar] [CrossRef] [PubMed]
- Guillaumet-Adkins, A.; Yañez, Y.; Peris-Diaz, M.D.; Calabria, I.; Palanca-Ballester, C.; Sandoval, J. Epigenetics and Oxidative Stress in Aging. Oxid. Med. Cell. Longev. 2017, 2017, 9175806. [Google Scholar] [CrossRef] [PubMed]
- Maynard, S.; Fang, E.F.; Scheibye-Knudsen, M.; Croteau, D.L.; Bohr, V.A. DNA Damage, DNA Repair, Aging, and Neurodegeneration. Cold Spring Harb. Perspect. Med. 2015, 5, a025130. [Google Scholar] [CrossRef] [PubMed]
- Gu, X.; Sun, J.; Li, S.; Wu, X.; Li, L. Oxidative stress induces DNA demethylation and histone acetylation in SH-SY5Y cells: Potential epigenetic mechanisms in gene transcription in Aβ production. Neurobiol. Aging 2013, 34, 1069–1079. [Google Scholar] [CrossRef]
- Bai, R.; Guo, J.; Ye, X.Y.; Xie, Y.; Xie, T. Oxidative stress: The core pathogenesis and mechanism of Alzheimer’s disease. Ageing Res. Rev. 2022, 77, 101619. [Google Scholar] [CrossRef]
- Quiroz-Baez, R.; Rojas, E.; Arias, C. Oxidative stress promotes JNK-dependent amyloidogenic processing of normally expressed human APP by differential modification of α-, β- and γ-secretase expression. Neurochem. Int. 2009, 55, 662–670. [Google Scholar] [CrossRef]
- Liu, Z.; Li, T.; Li, P.; Wei, N.; Zhao, Z.; Liang, H.; Ji, X.; Chen, W.; Xue, M.; Wei, J. The Ambiguous Relationship of Oxidative Stress, Tau Hyperphosphorylation, and Autophagy Dysfunction in Alzheimer’s Disease. Oxid. Med. Cell. Longev. 2015, 2015, 352723. [Google Scholar] [CrossRef]
- Su, B.; Wang, X.; Lee, H.G.; Tabaton, M.; Perry, G.; Smith, M.A.; Zhu, X. Chronic oxidative stress causes increased tau phosphorylation in M17 neuroblastoma cells. Neurosci. Lett. 2010, 468, 267–271. [Google Scholar] [CrossRef]
- Ionescu-Tucker, A.; Cotman, C.W. Emerging roles of oxidative stress in brain aging and Alzheimer’s disease. Neurobiol. Aging 2021, 107, 86–95. [Google Scholar] [CrossRef]
- Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Wilson, R.S. Vitamin E and cognitive decline in older persons. Arch. Neurol. 2002, 59, 1125–1132. [Google Scholar] [CrossRef]
- Kryscio, R.J.; Abner, E.L.; Caban-Holt, A.; Lovell, M.; Goodman, P.; Darke, A.K.; Yee, M.; Crowley, J.; Schmitt, F.A. Association of Antioxidant Supplement Use and Dementia in the Prevention of Alzheimer’s Disease by Vitamin E and Selenium Trial (PREADViSE). JAMA Neurol. 2017, 74, 567–573. [Google Scholar] [CrossRef]
- Mecocci, P.; Polidori, M.C. Antioxidant clinical trials in mild cognitive impairment and Alzheimer’s disease. Biochim. Biophys. Acta 2012, 1822, 631–638. [Google Scholar] [CrossRef]
- Sarti, P.; Forte, E.; Mastronicola, D.; Giuffrè, A.; Arese, M. Cytochrome c oxidase and nitric oxide in action: Molecular mechanisms and pathophysiological implications. Biochim. Biophys. Acta 2012, 1817, 610–619. [Google Scholar] [CrossRef]
- Lane, N. Cell biology: Power games. Nature 2006, 443, 901–903. [Google Scholar] [CrossRef]
- Barolet, D.; Christiaens, F.; Hamblin, M.R. Infrared and skin: Friend or foe. J. Photochem. Photobiol. B 2016, 155, 78–85. [Google Scholar] [CrossRef]
- Yang, L.; Youngblood, H.; Wu, C.; Zhang, Q. Mitochondria as a target for neuroprotection: Role of methylene blue and photobiomodulation. Transl. Neurodegener. 2020, 9, 19. [Google Scholar] [CrossRef]
- Amaroli, A.; Pasquale, C.; Zekiy, A.; Utyuzh, A.; Benedicenti, S.; Signore, A.; Ravera, S. Photobiomodulation and Oxidative Stress: 980 nm Diode Laser Light Regulates Mitochondrial Activity and Reactive Oxygen Species Production. Oxid. Med. Cell. Longev. 2021, 2021, 6626286. [Google Scholar] [CrossRef]
- Silveira, P.C.L.; Ferreira, G.K.; Zaccaron, R.P.; Glaser, V.; Remor, A.P.; Mendes, C.; Pinho, R.A.; Latini, A. Effects of photobiomodulation on mitochondria of brain, muscle, and C6 astroglioma cells. Med. Eng. Phys. 2019, 71, 108–113. [Google Scholar] [CrossRef]
- DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139, 136–153. [Google Scholar] [CrossRef]
- Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef]
- Rajesh, Y.; Kanneganti, T.D. Innate Immune Cell Death in Neuroinflammation and Alzheimer’s Disease. Cells 2022, 11, 1885. [Google Scholar] [CrossRef] [PubMed]
- Li, D.; Wu, M. Pattern recognition receptors in health and diseases. Signal Transduct. Target. Ther. 2021, 6, 291. [Google Scholar] [CrossRef] [PubMed]
- Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [PubMed]
- Shaftel, S.S.; Carlson, T.J.; Olschowka, J.A.; Kyrkanides, S.; Matousek, S.B.; O’Banion, M.K. Chronic interleukin-1β expression in mouse brain leads to leukocyte infiltration and neutrophil-independent blood brain barrier permeability without overt neurodegeneration. J. Neurosci. 2007, 27, 9301–9309. [Google Scholar] [CrossRef] [PubMed]
- Karki, R.; Sharma, B.R.; Tuladhar, S.; Williams, E.P.; Zalduondo, L.; Samir, P.; Zheng, M.; Sundaram, B.; Banoth, B.; Malireddi, R.K.S. Synergism of TNF-α and -γ Triggers Inflammatory Cell Death, Tissue Damage, and Mortality in SARS-CoV-2 Infection and Cytokine Shock Syndromes. Cell 2021, 184, 149–168.e17. [Google Scholar] [CrossRef]
- Karki, R.; Kanneganti, T.D. The ‘cytokine storm’: Molecular mechanisms and therapeutic prospects. Trends Immunol. 2021, 42, 681–705. [Google Scholar] [CrossRef]
- Savarin-Vuaillat, C.; Ransohoff, R.M. Chemokines and chemokine receptors in neurological disease: Raise, retain, or reduce? Neurotherapeutics 2007, 4, 590–601. [Google Scholar] [CrossRef]
- Xia, M.Q.; Qin, S.X.; Wu, L.J.; Mackay, C.R.; Hyman, B.T. Immunohistochemical study of the β-chemokine receptors CCR3 and CCR5 and their ligands in normal and Alzheimer’s disease brains. Am. J. Pathol. 1998, 153, 31–37. [Google Scholar] [CrossRef]
- Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
- Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, A.E.; Stevens, B.; Barres, B.A. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
- Sochocka, M.; Diniz, B.S.; Leszek, J. Inflammatory Response in the CNS: Friend or Foe? Mol. Neurobiol. 2017, 54, 8071–8089. [Google Scholar] [CrossRef]
- Wendiu, M.Y.; Hooks, S.B. Microglia phenotypes in aging and neurodegenerative diseases. Cells 2022, 11, 2091. [Google Scholar] [CrossRef]
- Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 2017, 169, 1276–1290.e17. [Google Scholar] [CrossRef]
- Mathys, H.; Adaikkan, C.; Gao, F.; Young, J.Z.; Manet, E.; Hemberg, M.; de Jager, P.L.; Ransohoff, R.M.; Regev, A.; Tsai, L.H. Temporal Tracking of Microglia Activation in Neurodegeneration at Single-Cell Resolution. Cell Rep. 2017, 21, 366–380. [Google Scholar] [CrossRef]
- Tan, Y.L.; Yuan, Y.; Tian, L. Microglial regional heterogeneity and its role in the brain. Mol. Psychiatry 2020, 25, 351–367. [Google Scholar] [CrossRef]
- Koenigsknecht, J.; Landreth, G. Microglial phagocytosis of fibrillar β-amyloid through a β1 integrin-dependent mechanism. J. Neurosci. 2004, 24, 9838–9846. [Google Scholar] [CrossRef]
- Alawieyah Syed Mortadza, S.; Sim, J.A.; Neubrand, V.E.; Jiang, L.H. A critical role of TRPM2 channel in Aβ42 -induced microglial activation and generation of tumor necrosis factor-α. Glia 2018, 66, 562–575. [Google Scholar] [CrossRef]
- White, C.S.; Lawrence, C.B.; Brough, D.; Rivers-Auty, J. Inflammasomes as therapeutic targets for Alzheimer’s disease. Brain Pathol. 2017, 27, 223–234. [Google Scholar] [CrossRef]
- Hawcroft, G.; Gardner, S.H.; Hull, M.A. Activation of peroxisome proliferator-activated receptor γ does not explain the antiproliferative activity of the nonsteroidal anti-inflammatory drug indomethacin on human colorectal cancer cells. J. Pharmacol. Exp. Ther. 2003, 305, 632–637. [Google Scholar] [CrossRef]
- Venegas, C.; Kumar, S.; Franklin, B.S.; Dierkes, T.; Brinkschulte, R.; Tejera, D.; Viera-Saecker, A.; Schwartz, S.; Santarelli, F.; Kummer, M.P.; et al. Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer’s disease. Nature 2017, 552, 355–361. [Google Scholar] [CrossRef]
- Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kügler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef] [PubMed]
- Hamelin, L.; Lagarde, J.; Dorothée, G.; Leroy, C.; Labit, M.; Comley, R.A.; de Souza, L.C.; Corne, H.; Dauphinot, L.; Bertoux, M.; et al. Early and protective microglial activation in Alzheimer’s disease: A prospective study using 18F-DPA-714 PET imaging. Brain 2016, 139, 1252–1264. [Google Scholar] [CrossRef] [PubMed]
- Bradburn, S.; Murgatroyd, C.; Ray, N. Neuroinflammation in mild cognitive impairment and Alzheimer’s disease: A meta-analysis. Ageing Res. Rev. 2019, 50, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Parhizkar, S.; Holtzman, D.M. APOE mediated neuroinflammation and neurodegeneration in Alzheimer’s disease. Semin. Immunol. 2022, 59, 101594. [Google Scholar] [CrossRef] [PubMed]
- Strittmatter, W.J.; Saunders, A.M.; Schmechel, D.; Pericak-Vance, M.; Enghild, J.; Salvesen, G.S.; Roses, A.D. Apolipoprotein E: High-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 1977–1981. [Google Scholar] [CrossRef]
- Boyles, J.K.; Pitas, R.E.; Wilson, E.; Mahley, R.W.; Taylor, J.M. Apolipoprotein E associated with astrocytic glia of the central nervous system and with nonmyelinating glia of the peripheral nervous system. J. Clin. Investig. 1985, 76, 1501–1513. [Google Scholar] [CrossRef]
- Lin, Y.T.; Seo, J.; Gao, F.; Feldman, H.M.; Wen, H.L.; Penney, J.; Cam, H.P.; Gjoneska, E.; Raja, W.K.; Cheng, J.; et al. APOE4 Causes Widespread Molecular and Cellular Alterations Associated with Alzheimer’s Disease Phenotypes in Human iPSC-Derived Brain Cell Types. Neuron 2018, 98, 1141–1154.e7. [Google Scholar] [CrossRef]
- Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic analysis of reactive astrogliosis. J. Neurosci. 2012, 32, 6391–6410. [Google Scholar] [CrossRef]
- Sienski, G.; Narayan, P.; Bonner, J.M.; Kory, N.; Boland, S.; Arczewska, A.A.; Ralvenius, W.T.; Akay, L.; Lockshin, E.; He, L. APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci. Transl. Med. 2021, 13, eaaz4564. [Google Scholar] [CrossRef]
- Tcw, J.; Qian, L.; Pipalia, N.H.; Chao, M.J.; Liang, S.A.; Shi, Y.; Jain, B.R.; Bertelsen, S.E.; Kapoor, M.; Marcora, E.; et al. Cholesterol and matrisome pathways dysregulated in astrocytes and microglia. Cell 2022, 185, 2213–2233.e25. [Google Scholar] [CrossRef]
- Ma, Y.; Li, P.; Ju, C.; Zuo, X.; Li, X.; Ding, T.; Liang, Z.; Zhang, J.; Li, K.; Wang, X.; et al. Photobiomodulation Attenuates Neurotoxic Polarization of Macrophages by Inhibiting the Notch1-HIF-1α/NF-κB Signalling Pathway in Mice with Spinal Cord Injury. Front. Immunol. 2022, 13, 816952. [Google Scholar] [CrossRef]
- Kigerl, K.A.; Gensel, J.C.; Ankeny, D.P.; Alexander, J.K.; Donnelly, D.J.; Popovich, P.G. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J. Neurosci. 2009, 29, 13435–13444. [Google Scholar] [CrossRef]
- Lu, Y.; Wang, R.; Dong, Y.; Tucker, D.; Zhao, N.; Ahmed, M.E.; Zhu, L.; Liu, T.C.; Cohen, R.M.; Zhang, Q. Low-level laser therapy for beta amyloid toxicity in rat hippocampus. Neurobiol. Aging 2017, 49, 165–182. [Google Scholar] [CrossRef]
- Shetty, A.K.; Zanirati, G. The Interstitial System of the Brain in Health and Disease. Aging Dis. 2020, 11, 200–211. [Google Scholar] [CrossRef]
- Kaiser, K.; Bryja, V. Choroid Plexus: The Orchestrator of Long-Range Signalling within the CNS. Int. J. Mol. Sci. 2020, 21, 4760. [Google Scholar] [CrossRef]
- Iliff, J.J.; Wang, M.; Liao, Y.; Plogg, B.A.; Peng, W.; Gundersen, G.A.; Benveniste, H.; Vates, G.E.; Deane, R.; Goldman, S.A.; et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 2012, 4, 147ra111. [Google Scholar] [CrossRef]
- Thrane, V.; Thrane, A.; Plog, B.; Thiyagarajan, M.; Iliff, J.J.; Deane, R.; Nagelhus, E.A.; Nedergaard, M. Paravascular microcirculation facilitates rapid lipid transport and astrocyte signaling in the brain. Sci. Rep. 2013, 3, 2582. [Google Scholar] [CrossRef]
- Silva, I.; Silva, J.; Ferreira, R.; Trigo, D. Glymphatic system, AQP4, and their implications in Alzheimer’s disease. Neurol. Res. Pract. 2021, 3, 5. [Google Scholar] [CrossRef]
- Meli, R.; Pirozzi, C.; Pelagalli, A. New Perspectives on the Potential Role of Aquaporins (AQPs) in the Physiology of Inflammation. Front. Physiol. 2018, 9, 101. [Google Scholar] [CrossRef]
- Nakada, T.; Kwee, I.L.; Igarashi, H.; Suzuki, Y. Aquaporin-4 Functionality and Virchow-Robin Space Water Dynamics: Physiological Model for Neurovascular Coupling and Glymphatic Flow. Int. J. Mol. Sci. 2017, 18, 1798. [Google Scholar] [CrossRef]
- Batiuk, M.Y.; Martirosyan, A.; Wahis, J.; de Vin, F.; Marneffe, C.; Kusserow, C.; Koeppen, J.; Viana, J.F.; Oliveira, J.F.; Voet, T.; et al. Identification of region-specific astrocyte subtypes at single cell resolution. Nat. Commun. 2020, 11, 1220. [Google Scholar] [CrossRef] [PubMed]
- Zeppenfeld, D.M.; Simon, M.; Haswell, J.D.; D’Abreo, D.; Murchison, C.; Quinn, J.F.; Grafe, M.R.; Woltjer, R.L.; Kaye, J.; Iliff, J.J. Association of Perivascular Localization of Aquaporin-4 With Cognition and Alzheimer Disease in Aging Brains. JAMA Neurol. 2017, 74, 91–99. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Lunde, L.K.; Nuntagij, P.; Oguchi, T.; Camassa, L.M.A.; Nilsson, L.N.G.; Lannfelt, L.; Xu, Y.; Amiry-Moghaddam, M.; Ottersen, O.P.; et al. Loss of astrocyte polarization in the tg-ArcSwe mouse model of Alzheimer’s disease. J. Alzheimers Dis. 2011, 27, 711–722. [Google Scholar] [CrossRef] [PubMed]
- Goedert, M.; Eisenberg, D.S.; Crowther, R.A. Propagation of Tau Aggregates and Neurodegeneration. Annu. Rev. Neurosci. 2017, 40, 189–210. [Google Scholar] [CrossRef]
- Harrison, I.F.; Ismail, O.; Machhada, A.; Colgan, N.; Ohene, Y.; Nahavandi, P.; Ahmed, Z.; Fisher, A.; Meftah, S.; Murray, T.K.; et al. Impaired glymphatic function and clearance of tau in an Alzheimer’s disease model. Brain 2020, 143, 2576–2593. [Google Scholar] [CrossRef]
- da Mesquita, S.; Louveau, A.; Vaccari, A.; Smirnov, I.; Cornelison, R.C.; Kingsmore, K.M.; Contarino, C.; Onengut-Gumuscu, S.; Farber, E.; Raper, D.; et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 2018, 560, 185–191. [Google Scholar] [CrossRef]
- Mogensen, F.L.; Delle, C.; Nedergaard, M. The Glymphatic System (En)during Inflammation. Int. J. Mol. Sci. 2021, 22, 7491. [Google Scholar] [CrossRef]
- Feng, W.; Zhang, Y.; Wang, Z.; Xu, H.; Wu, T.; Marshall, C.; Gao, J.; Xiao, M. Microglia prevent beta-amyloid plaque formation in the early stage of an Alzheimer’s disease mouse model with suppression of glymphatic clearance. Alzheimers Res. Ther. 2020, 12, 125. [Google Scholar] [CrossRef]
- Salehpour, F.; Khademi, M.; Bragin, D.E.; DiDuro, J.O. Photobiomodulation Therapy and the Glymphatic System: Promising Applications for Augmenting the Brain Lymphatic Drainage System. Int. J. Mol. Sci. 2022, 23, 2975. [Google Scholar] [CrossRef]
- Zinchenko, E.; Navolokin, N.; Shirokov, A.; Khlebtsov, B.; Dubrovsky, A.; Saranceva, E.; Abdurashitov, A.; Khorovodov, A.; Terskov, A.; Mamedova, A.; et al. Pilot study of transcranial photobiomodulation of lymphatic clearance of beta-amyloid from the mouse brain: Breakthrough strategies for non-pharmacologic therapy of Alzheimer’s disease. Biomed. Opt. Express 2019, 10, 4003–4017. [Google Scholar] [CrossRef]
- Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Dubrovsky, A.; Klimova, M.; Agranovich, I.; Terskov, A.; Shirokov, A.; Vinnik, V.; Kuzmina, A.; Lezhnev, N.; et al. Photobiomodulation of lymphatic drainage and clearance: Perspective strategy for augmentation of meningeal lymphatic functions. Biomed. Opt. Express 2020, 11, 725–734. [Google Scholar] [CrossRef]
- Lubart, R.; Eichler, M.; Lavi, R.; Friedman, H.; Shainberg, A. Low-energy laser irradiation promotes cellular redox activity. Photomed. Laser Surg. 2005, 23, 3–9. [Google Scholar] [CrossRef]
- Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Klimova, M.; Dubrovsky, A.; Shirokov, A.; Fomin, A.; Terskov, A.; Agranovich, I.; Mamedova, A.; Khorovodov, A.; et al. Photostimulation of cerebral and peripheral lymphatic functions. Transl. Biophotonics 2020, 2, e201900036. [Google Scholar] [CrossRef]
- Yue, X.; Mei, Y.; Zhang, Y.; Tong, Z.; Cui, D.; Yang, J.; Wang, A.; Wang, R.; Fei, X.; Ai, L.; et al. New insight into Alzheimer’s disease: Light reverses Aβ-obstructed interstitial fluid flow and ameliorates memory decline in APP/PS1 mice. Alzheimers Dement. 2019, 5, 671–684. [Google Scholar] [CrossRef]
- Sun, B.L.; Wang, L.H.; Yang, T.; Sun, J.Y.; Mao, L.L.; Yang, M.F.; Yuan, H.; Colvin, R.A.; Yang, X.Y. Lymphatic drainage system of the brain: A novel target for intervention of neurological diseases. Prog. Neurobiol. 2018, 163–164, 118–143. [Google Scholar] [CrossRef]
- Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 2011, 12, 723–738. [Google Scholar] [CrossRef]
- Soto-Rojas, L.O.; Cordoba, B.B.; Harrington, C.R.; Casas, A.S.; Alejandro, M.H.; Fierro, I.V.; Herrero, M.P.; Luna-Munoz, J. Insoluble Vascular Amyloid Deposits Trigger Disruption of the Neurovascular Unit in Alzheimer’s Disease Brains. J. Int. J. Mol. Sci. 2021, 22, 3654. [Google Scholar] [CrossRef]
- Iadecola, C. Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat. Rev. Neurosci. 2004, 5, 347–360. [Google Scholar] [CrossRef]
- Kisler, K.; Nelson, A.R.; Montagne, A.; Zlokovic, B.V. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat. Rev. Neurosci. 2017, 18, 419–434. [Google Scholar] [CrossRef]
- Mattsson, N.; Tosun, D.; Insel, P.S.; Simonson, A.; Jack, C.R., Jr.; Beckett, L.A.; Donohue, M.; Jagust, W.; Schuff, N.; Weiner, M.W.; et al. Association of brain amyloid-β with cerebral perfusion and structure in Alzheimer’s disease and mild cognitive impairment. Brain 2014, 137, 1550–1561. [Google Scholar] [CrossRef]
- Iturria-Medina, Y.; Sotero, R.C.; Toussaint, P.J.; Mateos-Pérez, J.M.; Evans, A.C.; Alzheimer’s Disease Neuroimaging Initiative. Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat. Commun. 2016, 7, 11934. [Google Scholar] [CrossRef] [PubMed]
- Smith, G.S.; de Leon, M.J.; George, A.E.; Kluger, A.; Volkow, N.D.; McRae, T.; Golomb, J.; Ferris, S.H.; Reisberg, B.; Ciaravino, J.; et al. Topography of cross-sectional and longitudinal glucose metabolic deficits in Alzheimer’s disease. Pathophysiologic implications. Arch. Neurol. 1992, 49, 1142–1150. [Google Scholar] [CrossRef]
- Kimura, T.; Hashimura, T.; Miyakawa, T. Observations of microvessels in the brain with Alzheimer’s disease by the scanning electron microscopy. Jpn. J. Psychiatry Neurol. 1991, 45, 671–676. [Google Scholar] [CrossRef] [PubMed]
- Nielsen, R.B.; Egefjord, L.; Angleys, H.; Mouridsen, K.; Geijl, M.; Møller, A.; Brock, B.; Brændgaard, H.; Gottrup, H.; Rungby, J.; et al. Capillary dysfunction is associated with symptom severity and neurodegeneration in Alzheimer’s disease. Alzheimers Dement. 2017, 13, 1143–1153. [Google Scholar] [CrossRef] [PubMed]
- Nortley, R.; Korte, N.; Izquierdo, P.; Hirunpattarasilp, C.; Mishra, A.; Jaunmuktane, Z.; Kyrargyri, V.; Pfeiffer, T.; Khennouf, L.; Madry, C.; et al. Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 2019, 365, eaav9518. [Google Scholar] [CrossRef]
- Korte, N.; Nortley, R.; Attwell, D. Cerebral blood flow decrease as an early pathological mechanism in Alzheimer’s disease. Acta Neuropathol. 2020, 140, 793–810. [Google Scholar] [CrossRef]
- Park, L.; Uekawa, K.; Garcia-Bonilla, L.; Koizumi, K.; Murphy, M.; Pistik, R.; Younkin, L.; Younkin, S.; Zhou, P.; Carlson, G.; et al. Brain Perivascular Macrophages Initiate the Neurovascular Dysfunction of Alzheimer Aβ Peptides. Circ. Res. 2017, 121, 258–269. [Google Scholar] [CrossRef]
- Palmer, J.C.; Barker, R.; Kehoe, P.G.; Love, S. Endothelin-1 is elevated in Alzheimer’s disease and upregulated by amyloid-β. J. Alzheimers Dis. 2012, 29, 853–861. [Google Scholar] [CrossRef]
- Murray, K.N.; Girard, S.; Holmes, W.M.; Parkes, L.M.; Williams, S.R.; Parry-Jones, A.R.; Allan, S.M. Systemic inflammation impairs tissue reperfusion through endothelin-dependent mechanisms in cerebral ischemia. Stroke 2014, 45, 3412–3419. [Google Scholar] [CrossRef]
- Cruz Hernández, J.C.; Bracko, O.; Kersbergen, C.J.; Muse, V.; Haft-Javaherian, M.; Berg, M.; Park, L.; Vinarcsik, L.K.; Ivasyk, I.; Rivera, D.A.; et al. Neutrophil adhesion in brain capillaries reduces cortical blood flow and impairs memory function in Alzheimer’s disease mouse models. Nat. Neurosci. 2019, 22, 413–420. [Google Scholar] [CrossRef]
- Cortes-Canteli, M.; Kruyer, A.; Fernandez-Nueda, I.; Marcos-Diaz, A.; Ceron, C.; Richards, A.T.; Jno-Charles, O.C.; Rodriguez, I.; Callejas, S.; Norris, E.H.; et al. Long-Term Dabigatran Treatment Delays Alzheimer’s Disease Pathogenesis in the TgCRND8 Mouse Model. J. Am. Coll. Cardiol. 2019, 74, 1910–1923. [Google Scholar] [CrossRef]
- Candelario-Jalil, E.; Dijkhuizen, R.M.; Magnus, T. Neuroinflammation, Stroke, Blood-Brain Barrier Dysfunction, and Imaging Modalities. Stroke 2022, 53, 1473–1486. [Google Scholar] [CrossRef]
- Brkic, M.; Balusu, S.; van Wonterghem, E.; Gorlé, N.; Benilova, I.; Kremer, A.; van Hove, I.; Moons, L.; de Strooper, B.; Kanazir, S.; et al. Amyloid β Oligomers Disrupt Blood-CSF Barrier Integrity by Activating Matrix Metalloproteinases. J. Neurosci. 2015, 35, 12766–12778. [Google Scholar] [CrossRef]
- Sun, X.; He, G.; Qing, H.; Zhou, W.; Dobie, F.; Cai, F.; Staufenbiel, M.; Huang, L.E.; Song, W. Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc. Natl. Acad. Sci. USA 2006, 103, 18727–18732. [Google Scholar] [CrossRef]
- Liu, S.L.; Wang, C.; Jiang, T.; Tan, L.; Xing, A.; Yu, J.T. The Role of Cdk5 in Alzheimer’s Disease. Mol. Neurobiol. 2016, 53, 4328–4342. [Google Scholar] [CrossRef]
- Mottet, D.; Dumont, V.; Deccache, Y.; Demazy, C.; Ninane, N.; Raes, M.; Michiels, C. Regulation of hypoxia-inducible factor-1α protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3β pathway in HepG2 cells. J. Biol. Chem. 2003, 278, 31277–31285. [Google Scholar] [CrossRef]
- Baik, J.S.; Lee, T.Y.; Kim, N.G.; Pak, K.; Ko, S.H.; Min, J.H.; Shin, Y. Effects of Photobiomodulation on Changes in Cognitive Function and Regional Cerebral Blood Flow in Patients with Mild Cognitive Impairment: A Pilot Uncontrolled Trial. J. Alzheimers Dis. 2021, 83, 1513–1519. [Google Scholar] [CrossRef]
- Chen, C.H.; Hung, H.S.; Hsu, S.H. Low-energy laser irradiation increases endothelial cell proliferation, migration, and eNOS gene expression possibly via PI3K signal pathway. Lasers Surg. Med. 2008, 40, 46–54. [Google Scholar] [CrossRef]
- Boelens, R.; Wever, R.; van Gelder, B.F.; Rademaker, H. An EPR study of the photodissociation reactions of oxidised cytochrome c oxidase-nitric oxide complexes. Biochim. Biophys. Acta 1983, 724, 176–183. [Google Scholar] [CrossRef]
- Keszler, A.; Brandal, G.; Baumgardt, S.; Ge, Z.D.; Pratt, P.F.; Riess, M.L.; Bienengraeber, M. Far red/near infrared light-induced protection against cardiac ischemia and reperfusion injury remains intact under diabetic conditions and is independent of nitric oxide synthase. Front. Physiol. 2014, 5, 305. [Google Scholar] [CrossRef]
- Yokomizo, S.; Roessing, M.; Morita, A.; Kopp, T.; Ogawa, E.; Katagiri, W.; Feil, S.; Huang, P.L.; Atochin, D.N.; Kashiwagi, S. Near-infrared II photobiomodulation augments nitric oxide bioavailability via phosphorylation of endothelial nitric oxide synthase. FASEB J. 2022, 36, e22490. [Google Scholar] [CrossRef] [PubMed]
- Cury, V.; Moretti, A.I.; Assis, L.; Bossini, P.; Crusca, J.d.S.; Neto, C.B.; Fangel, R.; de Souza, H.P.; Hamblin, M.R.; Parizotto, N.A. Low level laser therapy increases angiogenesis in a model of ischemic skin flap in rats mediated by VEGF, HIF-1α and MMP-2. J. Photochem. Photobiol. B 2013, 125, 164–170. [Google Scholar] [CrossRef] [PubMed]
- Mullard, A. Landmark Alzheimer’s Drug Approval Confounds Research Community. Available online: https://www.nature.com/articles/d41586-021-01546-2 (accessed on 20 March 2023).
- Budd Haeberlein, S.; Aisen, P.S.; Barkhof, F.; Chalkias, S.; Chen, T.; Cohen, S.; Dent, G.; Hansson, O.; Harrison, K.; von Hehn, C. Two Randomized Phase 3 Studies of Aducanumab in Early Alzheimer’s Disease. J. Prev. Alzheimers Dis. 2022, 9, 197–210. [Google Scholar] [CrossRef] [PubMed]
- Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef]
- Albrecht, D.S.; Sagare, A.; Pachicano, M.; Sweeney, M.D.; Toga, A.; Zlokovic, B.; Chui, H.; Joe, E.; Schneider, L.; Morris, J.C.; et al. Early neuroinflammation is associated with lower amyloid and tau levels in cognitively normal older adults. Brain Behav. Immun. 2021, 94, 299–307. [Google Scholar] [CrossRef]
- Leng, F.; Hinz, R.; Gentleman, S.; Hampshire, A.; Dani, M.; Brooks, D.J.; Edison, P. Neuroinflammation is independently associated with brain network dysfunction in Alzheimer’s disease. Mol. Psychiatry 2023, 28, 1303–1311. [Google Scholar] [CrossRef]
- Takeuchi, H.; Mizuno, T.; Zhang, G.; Wang, J.; Kawanokuchi, J.; Kuno, R.; Suzumura, A. Neuritic beading induced by activated microglia is an early feature of neuronal dysfunction toward neuronal death by inhibition of mitochondrial respiration and axonal transport. J. Biol. Chem. 2005, 280, 10444–10454. [Google Scholar] [CrossRef]
- Villegas-Llerena, C.; Phillips, A.; Garcia-Reitboeck, P.; Hardy, J.; Pocock, J.M. Microglial genes regulating neuroinflammation in the progression of Alzheimer’s disease. Curr. Opin. Neurobiol. 2016, 36, 74–81. [Google Scholar] [CrossRef]
- Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
- Zhang, Y.; Chen, K.; Sloan, S.A.; Bennett, M.L.; Scholze, A.R.; O’Keeffe, S.; Phatnani, H.P.; Guarnieri, P.; Caneda, C.; Ruderisch, N.; et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 2014, 34, 11929–11947. [Google Scholar] [CrossRef]
- Shimohama, S.; Tanino, H.; Kawakami, N.; Okamura, N.; Kodama, H.; Yamaguchi, T.; Hayakawa, T.; Nunoura, A.; Chiba, S.; Perry, G.; et al. Activation of NADPH oxidase in Alzheimer’s disease brains. Biochem. Biophys. Res. Commun. 2000, 273, 5–9. [Google Scholar] [CrossRef]
- Geng, L.; Fan, L.M.; Liu, F.; Smith, C.; Li, J.M. Nox2 dependent redox-regulation of microglial response to amyloid-β stimulation and microgliosis in aging. Sci. Rep. 2020, 10, 1582. [Google Scholar] [CrossRef]
- Luo, J.; Mills, K.; le Cessie, S.; Noordam, R.; van Heemst, D. Ageing, age-related diseases and oxidative stress: What to do next? Ageing Res. Rev. 2020, 57, 100982. [Google Scholar] [CrossRef]
- Sharifi-Rad, M.; Anil Kumar, N.V.; Zucca, P.; Varoni, E.M.; Dini, L.; Panzarini, E.; Rajkovic, J.; Fokou, P.V.T.; Azzini, E.; Peluso, I.; et al. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front. Physiol. 2020, 11, 694. [Google Scholar] [CrossRef]
- Antunes Dos Santos, A.; Ferrer, B.; Marques Gonçalves, F.; Tsatsakis, A.M.; Renieri, E.A.; Skalny, A.V.; Farina, M.; Rocha, J.B.T.; Aschner, M. Oxidative Stress in Methylmercury-Induced Cell Toxicity. Toxics 2018, 6, 47. [Google Scholar] [CrossRef]
- Albargothy, N.J.; Johnston, D.A.; MacGregor-Sharp, M.; Weller, R.O.; Verma, A.; Hawkes, C.A.; Carare, R.O. Convective influx/glymphatic system: Tracers injected into the CSF enter and leave the brain along separate periarterial basement membrane pathways. Acta Neuropathol. 2018, 136, 139–152. [Google Scholar] [CrossRef]
Gene | Chr | Age of Onset (Form of AD) | Gene Function Linked to AD | Effects of Gene Mutations on AD Pathogenesis |
---|---|---|---|---|
APP | 21 | Early/(Familial) | Aβ production, neural survival, synaptic function | Increases Aβ accumulation |
PSEN1 | 14 | Early/(Familial) | Aβ production, γ-secretase activity, calcium signaling | Increases Aβ42/Aβ40 ratio |
PSEN2 | 1 | Early/(Familial) | Aβ production, γ-secretase activity, synaptic plasticity | Increases Aβ42/Aβ40 ratio |
APOE | 19 | Late/(Sporadic) | Lipid transport and metabolism, synaptic metabolism | Reduces Aβ clearance, increases Aβ deposition |
ABCA7 | 9 | Late/(Sporadic) | Phagocytosis, Aβ clearance | Increases Aβ accumulation |
CD33 | 19 | Late/(Sporadic) | Immune response, microglial phagocytosis | Increases Aβ accumulation, prevents Aβ from microglial clearance |
TREM2 | 6 | Late/(Sporadic) | Immune response, microglial phagocytosis | Reduces Aβ microglial clearance |
CLU | 8 | Late/(Sporadic) | Immune modulation, cell death regulation | Increases Aβ aggregation, reduces Aβ clearance |
SORL1 | 11 | Late/(Sporadic) | Lipid metabolism, APP trafficking | Reduces Aβ destruction and endosomal degradation, impairs APP processing |
CR1 | 1 | Late/(Sporadic) | Immune response, Aβ clearance | Reduces Aβ clearance |
MS4A | 11 | Late/(Sporadic) | Calcium signaling, immune function | Reduces sTREM2 and Aβ clearance |
BIN1 | 2 | Late/(Sporadic) | Endocytosis, cytoskeletal organization | Deregulates early endosome trafficking, induces neuron degeneration |
CD2AP | 6 | Late/(Sporadic) | Endocytosis, cell signaling, cytoskeletal organization | Increases Aβ plaque formation, synaptic dysfunction, neurotoxicity |
PICALM | 11 | Late/(Sporadic) | Endocytosis, synapse function, iron homeostasis | Increases Aβ production, reduces Aβ clearance, mediates tau neurodegeneration |
EPHA1 | 7 | Late/(Sporadic) | Immune response, synapse function, apoptosis | Promotes synaptic dysfunction and neurons apoptosis |
INPP5D | 2 | Late/(Sporadic) | Immune regulation, cytokine signaling | Increases plaque deposition, promotes microglial and synaptic dysfunction |
MEF2C | 5 | Late/(Sporadic) | Synapse construction, regulation of microglia | Mediates microglial overstimulation and increases neuroinflammation in late-onset |
CASS4 | 20 | Late/(Sporadic) | Cell migration and adhesion, inflammation | Increases NFT formation |
PTK2B | 8 | Late/(Sporadic) | Cell migration, adhesion, and proliferation | Modulates tau pathogenesis, increases NFT formation |
Biomarker | Importance | |
---|---|---|
Amyloid biomarkers | APP | APP cleavage by γ- and β-secretases results in Aβ formation |
Aβ42/Aβ40 | Reduced Aβ42/Aβ40 ratio is observed in AD | |
Tau biomarkers | T-tau | Elevated in prodromal and dementia AD |
P-tau | Hyperphosphorylation of tau leads to NFT formation Elevated in prodromal and dementia AD High P-tau in CSF is only observed in AD | |
Neural damage biomarkers | NfL | Marker for acute brain damage and neurodegeneration, but not specific for AD |
S100β | High levels correlate with greater brain atrophy | |
Neuroinflammation biomarkers | GFAP | Marker of astrocyte activation Observed to be higher in preclinical AD cases |
TNF-α | Pro-inflammatory cytokine frequently reported to be elevated in blood plasma and CSF of AD patients | |
IL-β | Promotes Aβ plaque and NFT formation | |
Synaptic biomarkers | α-synuclein | Elevated in CSF of MCI and AD patients |
Neurogranin | High neurogranin is observed in AD and reflects synaptic (dendritic) degeneration | |
Metabolic biomarkers | ApoE | Major lipid transporter in the brain May lead to synaptic defects and cognitive impairments |
GDNF | Promotes dopamine uptake in dopaminergic neurons Significant decrease reported in AD patients |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Su, M.; Nizamutdinov, D.; Liu, H.; Huang, J.H. Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 9272. https://doi.org/10.3390/ijms24119272
Su M, Nizamutdinov D, Liu H, Huang JH. Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer’s Disease. International Journal of Molecular Sciences. 2023; 24(11):9272. https://doi.org/10.3390/ijms24119272
Chicago/Turabian StyleSu, Matthew, Damir Nizamutdinov, Hanli Liu, and Jason H. Huang. 2023. "Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer’s Disease" International Journal of Molecular Sciences 24, no. 11: 9272. https://doi.org/10.3390/ijms24119272
APA StyleSu, M., Nizamutdinov, D., Liu, H., & Huang, J. H. (2023). Recent Mechanisms of Neurodegeneration and Photobiomodulation in the Context of Alzheimer’s Disease. International Journal of Molecular Sciences, 24(11), 9272. https://doi.org/10.3390/ijms24119272