Perspectives on the profound pathological mechanisms of Alzheimer's disease and the corresponding treatment strategies

LIN Yong, GU Ben J.

Chinese Journal of Alzheimer's Disease and Related Disorders ›› 2025, Vol. 8 ›› Issue (4) : 219-225.

PDF(984 KB)
Home Journals Chinese Journal of Alzheimer's Disease and Related Disorders
Chinese Journal of Alzheimer's Disease and Related Disorders

Abbreviation (ISO4): Chinese Journal of Alzheimer's Disease and Related Disorders      Editor in chief: Jun WANG

About  /  Aim & scope  /  Editorial board  /  Indexed  /  Contact  / 
PDF(984 KB)
Chinese Journal of Alzheimer's Disease and Related Disorders ›› 2025, Vol. 8 ›› Issue (4) : 219-225. DOI: 10.3969/j.issn.2096-5516.2025.04.001
Commentary

Perspectives on the profound pathological mechanisms of Alzheimer's disease and the corresponding treatment strategies

Author information +
History +

Abstract

There have been many different opinions on the pathogenesis of Alzheimer's disease (AD), and the β-amyloid protein (Aβ) theory has always been dominant. However, many treatments targeting on Aβ have had little effect, which has led researchers to re-examine this theory. More and more research findings have made people realize that although the accumulation of Aβ plaques in the brain is closely related to cognitive decline, it may only be a manifestation of AD, not the root cause of this disease. The body's inability to clear the accumulation of proteins including Aβ due to various reasons such as chronic inflammation with brain damage may be a deeper cause of this disease. These important cell functions include endocytosis, macropinocytosis, innate phagocytosis and autophagy, and mitochondria that provide energy to these high-energy consumption applications may be the determining factor affecting these functions. Therefore, simply removing Aβ plaques may have little effect in the treatment of Alzheimer's disease. More efforts should be placed on how to improve essential cell functions like innate phagocytosis and autophagy, as well as to improve mitochondrial function to increase energy production.

Key words

Alzheimer's disease / Aβ theory / Innate phagocytosis / Autophagy / Mitochondrial / Energy metabolism

Cite this article

Download Citations
LIN Yong , GU Ben J.. Perspectives on the profound pathological mechanisms of Alzheimer's disease and the corresponding treatment strategies[J]. Chinese Journal of Alzheimer's Disease and Related Disorders. 2025, 8(4): 219-225 https://doi.org/10.3969/j.issn.2096-5516.2025.04.001

References

[1]
Masters CL, Simms G, Weinman NA, et al. Amyloid plaque core protein in Alzheimer disease and down syndrome[J]. Proc Natl Acad Sci U S A, 1985, 82(12):4245-4249.
[2]
Masters CL, Multhaup G, Simms G, et al. Neuronal origin of a cerebral amyloid: neurofibrillary tangles of Alzheimer's disease contain the same protein as the amyloid of plaque cores and blood vessels[J]. EMBO J, 1985, 4(11):2757-2763.
[3]
Glenner GG, Wong CW. Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein[J]. Biochem Biophys Res Commun, 1984, 120(3):885-890.
[4]
Bancher C, Brunner C, Lassmann H, et al. Accumulation of abnormally phosphorylated tau precedes the formation of neurofibrillary tangles in Alzheimer's disease[J]. Brain Res, 1989, 477(1-2):90-99.
[5]
Chen GF, Xu TH, Yan Y, et al. Amyloid beta: structure, biology and structure-based therapeutic development[J]. Acta Pharmacol Sin, 2017, 38(9):1205-1235.
[6]
Chen QS, Kagan BL, Hirakura Y, et al. Impairment of hippocampal long-term potentiation by Alzheimer amyloid beta-peptides[J]. J Neurosci Res, 2000, 60(1):65-72.
[7]
Arbor SC, LaFontaine M, Cumbay M, et al. Amyloid-beta Alzheimer targets - protein processing, lipid rafts, and amyloid-beta pores[J]. Yale J Biol Med, 2016, 89(1):5-21.
[8]
Bernstein SL, Dupuis NF, Lazo ND, et al. Amyloid-beta protein oligomerization and the importance of tetramers and dodecamers in the aetiology of Alzheimer's disease[J]. Nat Chem, 2009, 1(4):326-331.
[9]
Thakur S, Dhapola R, Sarma P, et al. Neuroinflammation in Alzheimer's disease: current progress in molecular signaling and therapeutics[J]. Inflammation, 2023, 46(1):1-17.
[10]
Chen Z, Zhong C. Oxidative stress in Alzheimer's disease[J]. Neurosci Bull, 2014, 30(2):271-281.
[11]
Reinisalo M, Karlund A, Koskela A, et al. Polyphenol stilbenes: molecular mechanisms of defence against oxidative stress and aging-related diseases[J]. Oxid Med Cell Longev, 2015, 2015:340520.
[12]
Mutter J, Curth A, Naumann J, et al. Does inorganic mercury play a role in Alzheimer's disease? a systematic review and an integrated molecular mechanism[J]. J Alzheimers Dis, 2010, 22(2):357-374.
[13]
Rozga M, Bal W. The Cu(II)/Abeta/human serum albumin model of control mechanism for copper-related amyloid neurotoxicity[J]. Chem Res Toxicol, 2010, 23(2):298-308.
[14]
Lei P, Walker T, Ayton S, et al. Neuroferroptosis in health and diseases[J]. Nat Rev Neurosci, 2025.
[15]
Zhang X, Wang J, Zhang Z, et al. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies[J]. Transl Neurodegener, 2024, 13(1):40.
[16]
Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein-E Type-4 allele and the risk of Alzheimers-disease in late-onset families[J]. Science, 1993, 261(5123):921-923.
[17]
Liu S, Park S, Allington G, et al. Targeting apolipoprotein E/Amyloid beta binding by peptoid CPO_Abeta17-21 P ameliorates Alzheimer's disease related pathology and cognitive decline[J]. Sci Rep, 2017, 7(1):8009.
[18]
Garai K, Verghese PB, Baban B, et al. The binding of apolipoprotein E to oligomers and fibrils of amyloid-beta alters the kinetics of amyloid aggregation[J]. Biochemistry, 2014, 53(40):6323-6331.
[19]
Ruiz J, Kouiavskaia D, Migliorini M, et al. The apoE isoform binding properties of the VLDL receptor reveal marked differences from LRP and the LDL receptor[J]. J Lipid Res, 2005, 46(8):1721-1731.
[20]
Guo JL, Braun D, Fitzgerald GA, et al. Decreased lipidated ApoE-receptor interactions confer protection against pathogenicity of ApoE and its lipid cargoes in lysosomes[J]. Cell, 2025, 188(1):187-206.
[21]
Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis[J]. Biol Psychiatry, 2015, 77(1):43-51.
[22]
Kunkle BW, Grenier-Boley B, Sims R, et al. Genetic meta-analysis of diagnosed Alzheimer's disease identifies new risk loci and implicates abeta, tau, immunity and lipid processing[J]. Nat Genet, 2019, 51(3):414-430.
[23]
Jiao B, Xiao X, Yuan Z, et al. Associations of risk genes with onset age and plasma biomarkers of Alzheimer's disease: a large case-control study in mainland China[J]. Neuropsychopharmacology, 2022, 47(5):1121-1127.
[24]
Arndt JW, Qian F, Smith BA, et al. Structural and kinetic basis for the selectivity of aducanumab for aggregated forms of amyloid-beta[J]. Sci Rep, 2018, 8(1):6412.
[25]
Chad JS, Yong Z, Shobha D, et al. A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer's disease with lecanemab, an anti-Aβ protofibril antibody[J]. Alzheimers Res Ther, 2021, 13(1):80.
[26]
Minton MA, Lo AC, Evans CD, et al. Donanemab in early Alzheimer's disease[J]. New Engl J Med, 2021, 384(18):1691-1704.
[27]
Kim B-H, Kim S, Nam Y, et al. Second-generation anti-amyloid monoclonal antibodies for Alzheimer's disease: current landscape and future perspectives[J]. Transl Neurodegener, 2025, 14(1):6.
[28]
Robakis NK. Mechanisms of AD neurodegeneration may be independent of Aβ and its derivatives[J]. Neurobiol Aging, 2011, 32(3):372-379.
[29]
Kadamangudi S, Marcatti M, Zhang WR, et al. Amyloid-β oligomers increase the binding and internalization of tau oligomers in human synapses[J]. Acta Neuropathol, 2024, 149(1):2.
[30]
Wakeman DR, Weed MR, Perez SE, et al. Intrathecal amyloid-β oligomer administration increases tau phosphorylation in the medial temporal lobe in the African green monkey: a nonhuman primate model of Alzheimer's disease[J]. Neuropathol Appl Neurobiol, 2022, 48(4):e12800.
[31]
Yue F, Feng S, Lu C, et al. Synthetic amyloid-beta oligomers drive early pathological progression of Alzheimer's disease in nonhuman primates[J]. IScience, 2021, 24(10):103207.
[32]
Johnson LV, Leitner WP, Rivest AJ, et al. The Alzheimer's abeta-peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration[J]. Proc National Acad Sci U S A, 2002, 99(18):11830.
[33]
Mullins RF, Russell SR, Hageman GS, et al. Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease[J]. FASEB J, 2000, 14(7):835-846.
[34]
Vilchez D, Saez I, Dillin A, et al. The role of protein clearance mechanisms in organismal ageing and age-related diseases[J]. Nat Commun, 2014, 5:5659.
[35]
Zadka Ł, Sochocka M, Hachiya N, et al. Endocytosis and Alzheimer's disease[J]. GeroScience, 2024, 46(1):71-85.
[36]
Ando K, Brion JP, Stygelbout V, et al. Clathrin adaptor CALM/PICALM is associated with neurofibrillary tangles and is cleaved in Alzheimer's brains[J]. Acta Neuropathol, 2013, 125(6):861-878.
[37]
Park TE, Singh B, Li H, et al. Enhanced BBB permeability of osmotically active poly(mannitol-co-PEI) modified with rabies virus glycoprotein via selective stimulation of caveolar endocytosis for RNAi therapeutics in Alzheimer's disease[J]. Biomaterials, 2015, 38:61-71.
[38]
Wu J, Zhou SL, Pi LH, et al. High glucose induces formation of tau hyperphosphorylation via Cav-1-mTOR pathway: a potential molecular mechanism for diabetes-induced cognitive dysfunction[J]. Oncotarget, 2017, 8(25):40843-40856.
[39]
Mandrekar S, Jiang Q, Lee CYD, et al. Microglia mediate the clearance of soluble aβ through fluid phase macropinocytosis[J]. J Neurosci, 2009, 29(13):4252-4262.
[40]
Prabhudas M, Bowdish D, Drickamer K, et al. Standardizing scavenger receptor nomenclature[J]. J Immunol, 2014, 192(5):1997-2006.
[41]
Gu BJ, Saunders BM, Jursik C, et al. The P2X7-nonmuscle myosin membrane complex regulates phagocytosis of nonopsonized particles and bacteria by a pathway attenuated by extracellular ATP[J]. Blood, 2010, 115(8):1621-1631.
[42]
Gu BJ, Saunders BM, Petrou S, et al. P2X(7) is a scavenger receptor for apoptotic cells in the absence of its ligand, extracellular ATP[J]. J Immunol, 2011, 187(5):2365-2375.
[43]
Gu BJ, Rathsam C, Stokes L, et al. Extracellular ATP dissociates nonmuscle myosin from P2X(7) complex: this dissociation regulates P2X(7) pore formation[J]. Am J Physiol Cell Physiol, 2009, 297(2):C430-439.
[44]
Park YM, Drazba JA, Vasanji A, et al. Oxidized LDL/CD36 interaction induces loss of cell polarity and inhibits macrophage locomotion[J]. Mol Biol Cell, 2012, 23(16):3057-3068.
[45]
Gomez CP, Descoteaux A. Moesin and myosin IIA modulate phagolysosomal biogenesis in macrophages[J]. Biochem Biophys Res Commun, 2018, 495(2):1964-1971.
[46]
Rayaprolu S, Gao T, Xiao H, et al. Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer's disease[J]. Mol Neurodegener, 2020, 15(1):28.
[47]
Gu BJ, Wiley JS. P2X7 as a scavenger receptor for innate phagocytosis in the brain[J]. Br J Pharmacol, 2018, 175(22):4195-4208.
[48]
Gu BJ, Wiley JS. Broad applications of multi-colour time-resolved flow cytometry[C]. In: Schmid I, ed. Flow Cytometry: Recent Perspectives. Croatia; InTech 2012:185-202.
[49]
Gu BJ, Sun C, Fuller S, et al. A quantitative method for measuring innate phagocytosis by human monocytes using real-time flow cytometry[J]. Cytometry A, 2014, 85(4):313-321.
[50]
Gu BJ, Duce JA, Valova VA, et al. P2X7 receptor-mediated scavenger activity of mononuclear phagocytes toward non-opsonized particles and apoptotic cells is inhibited by serum glycoproteins but remains active in cerebrospinal fluid[J]. J Biol Chem, 2012, 287(21):17318-17330.
[51]
Lovelace MD, Gu BJ, Eamegdool SS, et al. P2X7 receptors mediate innate phagocytosis by human neural precursor cells and neuroblasts[J]. Stem Cells, 2015, 33(2):526-541.
[52]
Vessey KA, Gu BJ, Jobling AI, et al. Loss of Function of P2X7 receptor scavenger activity in aging mice: a novel model for investigating the early pathogenesis of age-related macular degeneration[J]. Am J Pathol, 2017, 187(8):1670-1685.
[53]
De Maeyer RPH, Chambers ES. The impact of ageing on monocytes and macrophages[J]. Immunol Lett, 2021, 230:1-10.
[54]
Hampton K, Hellmich C, Maynard RS, et al. LC3-Associated phagocytosis is impaired in bone marrow macrophages during ageing leading to defective processing of apoptotic cells and suppression of sting activation[J]. Blood, 2022, 140(Supplement 1):5759-5759.
[55]
Li W. Phagocyte dysfunction, tissue aging and degeneration[J]. Ageing Res Rev, 2013, 12(4):1005-1012.
[56]
Kumakura S, Yamaguchi Y, Murakawa Y, et al. Effect of cytokines and hyperthermia on phagocytosis and phosphatidylserine externalization: implication for the pathophysiology of hemophagocytic syndrome[J]. Ann Clin Lab Sci, 2018, 48(3):314-322.
[57]
Protsiv M, Ley C, Lankester J, et al. Decreasing human body temperature in the United States since the industrial revolution[J]. Elife, 2020, 9:e49555.
[58]
Wang J, Gu BJ, Masters CL, et al. A systemic view of Alzheimer disease - insights from amyloid-beta metabolism beyond the brain[J]. Nat Rev Neurol, 2017, 13(10):612-623.
[59]
Gu BJ, Huang X, Ou A, et al. Innate phagocytosis by peripheral blood monocytes is altered in Alzheimer's disease[J]. Acta Neuropathol, 2016, 132(3):377-389.
[60]
Huang X, Fowler C, Li Y, et al. Clearance and transport of amyloid beta by peripheral monocytes correlate with Alzheimer's disease progression[J]. Nat Commun, 2024, 15(1):7998.
[61]
Huang X, Li Y, Fowler C, et al. Leukocyte surface biomarkers implicate deficits of innate immunity in sporadic Alzheimer's disease[J]. Alzheimers Dement, 2022, 19(5):2084-2094.
[62]
Li Y, Huang X, Fowler C, et al. Identification of leukocyte surface P2X7 as a biomarker associated with Alzheimer's disease[J]. Int J Mol Sci, 2022, 23(14):7867.
[63]
Gu BJ, Sluyter R, Skarratt KK, et al. An Arg307 to gln polymorphism within the ATP-binding site causes loss of function of the human P2X7 receptor[J]. J Biol Chem, 2004, 279(30):31287-31295.
[64]
Gu BJ, Field J, Dutertre S, et al. A rare P2X7 variant Arg307Gln with absent pore formation function protects against neuroinflammation in multiple sclerosis[J]. Hum Mol Genet, 2015, 24(19):5644-5654.
[65]
Sadovnick AD, Gu BJ, Traboulsee AL, et al. Purinergic receptors P2RX4 and P2RX7 in familial multiple sclerosis[J]. Hum Mutat, 2017, 38(6):736-744.
[66]
Gu BJ, Baird PN, Vessey KA, et al. A rare functional haplotype of the P2RX4 and P2RX7 genes leads to loss of innate phagocytosis and confers increased risk of age-related macular degeneration[J]. FASEB J, 2013, 27(4):1479-1487.
[67]
Gu BJ, Huang X, Avula PK, et al. Deficits in monocyte function in age related macular degeneration: a novel systemic change associated with the disease[J]. Front Med, 2021, 8:634177.
[68]
Wiley JS, Gu BJ. A new role for the P2X7 receptor: a scavenger receptor for bacteria and apoptotic cells in the absence of serum and extracellular ATP[J]. Purinergic Signal, 2012, 8(3):579-586.
[69]
Leeson HC, Kasherman MA, Chan-Ling T, et al. P2X7 receptors regulate phagocytosis and proliferation in adult hippocampal and SVZ neural progenitor cells: implications for inflammation in neurogenesis[J]. Stem Cells, 2018, 36(11):1764-1777.
[70]
Hu Z, Yang B, Mo X, et al. Mechanism and regulation of autophagy and its role in neuronal diseases[J]. Mol Neurobiol, 2015, 52(3):1190-1209.
[71]
Giong HK, Hyeon SJ, Lee JG, et al. Tau accumulation is cleared by the induced expression of VCP via autophagy[J]. Acta Neuropathol, 2024, 148(1):46.
[72]
Gorantla NV, Chinnathambi S. Autophagic pathways to clear the tau aggregates in Alzheimer's disease[J]. Cell Mol Neurobiol, 2021, 41(6):1175-1181.
[73]
NKrance SH, Wu CY, Chan ACY, et al. Endosomal-lysosomal and autophagy pathway in Alzheimer's disease: a systematic review and meta-analysis[J]. J Alzheimers Dis, 2022, 88(4):1279-1292.
[74]
Kerr JS, Adriaanse BA, Greig NH, et al. Mitophagy and Alzheimer's disease: cellular and molecular mechanisms[J]. Trends Neurosci, 2017, 40(3):151-166.
[75]
Martínez RAS, Pinky PD, Harlan BA, et al. GTP energy dependence of endocytosis and autophagy in the aging brain and Alzheimer's disease[J]. GeroScience, 2023, 45(2):757-780.
[76]
MSilva DF, Esteves AR, Oliveira CR, et al. Mitochondrial metabolism power SIRT2-dependent deficient traffic causing Alzheimer's-disease related pathology[J]. Mol Neurobiol, 2017, 54(6):4021-4040.
[77]
Fairley LH, Kei Onn L, Jia Hui W, et al. Mitochondrial control of microglial phagocytosis by the translocator protein and hexokinase 2 in Alzheimer's disease[J]. Proc National Acad Sci U S A, 2023, 120(8):1-12.
[78]
Zhang H, Wang H, Gao F, et al. TSPO deficiency accelerates amyloid pathology and neuroinflammation by impairing microglial phagocytosis[J]. Neurobiol Aging, 2021, 106:292-303.
[79]
Jadala C, Robo MT, Richardson TI, et al. Exploring Class‐II PI3K inhibition for the treatment of Alzheimer's disease: virtual screening for PI3KC2A inhibitors[J]. Alzheimers Amp Dement, 2024, 20(S6):1-2.
[80]
Yoon Sun C, Oh-hoon K, Hyun Geun O, et al. Justicidin a reduces β-Amyloid via inhibiting endocytosis of β-Amyloid precursor protein[J]. Biomol Ther, 2019, 27(3):276-282.
[81]
Cole GM, Yang F, Lim GP, et al. A rationale for curcuminoids for the prevention or treatment of Alzheimer's disease[J]. Current Medicinal Chemistry - Immunology, Endocrine & Metabolic Agents, 2003, 3(1):15.
[82]
Mizwicki MT, Menegaz D, Zhang J, et al. Genomic and nongenomic signaling induced by 1α25(OH)2-Vitamin D3 promotes the recovery of Amyloid-β phagocytosis by Alzheimer's disease macrophages[J]. J Alzheimers Dis, 2012, 29(1):51-62.
[83]
Mizwicki MT, Liu G, Fiala M, et al. 1α25-Dihydroxyvitamin D3 and Resolvin D1 retune the balance between Amyloid-β phagocytosis and inflammation in Alzheimer's disease patients[J]. J Alzheimers Dis, 2013, 34(1):155-170.
[84]
Landgraf P, Wahle P, Pape HC, et al. The survival-promoting peptide Y-P30 enhances binding of pleiotrophin to syndecan-2 and -3 and supports its neuritogenic activity[J]. J Biol Chem, 2008, 283(36):25036-25045.
[85]
Bellizzi MJ, Hammond JW, Li H, et al. The mixed-lineage kinase inhibitor URMC-099 protects hippocampal synapses in experimental autoimmune encephalomyelitis[J]. eNeuro, 2018, 5(6): ENEURO.0245-18.2018.
[86]
Tajbakhsh A, Read M, Barreto GE, et al. Apoptotic neurons and amyloid-beta clearance by phagocytosis in Alzheimer's disease: pathological mechanisms and therapeutic outlooks[J]. Eur J Pharmacol, 2021, 895:173873.
[87]
Yang S, Du Y, Zhao X, et al. Cannabidiol enhances microglial beta-amyloid peptide phagocytosis and clearance via vanilloid family type 2 channel activation[J]. Int J Mol Sci, 2022, 23(10):5367.
[88]
Fernandes SM, Mayer J, Nilsson P, et al. How close is autophagy-targeting therapy for Alzheimer's disease to clinical use? a summary of autophagy modulators in clinical studies[J]. Front Cell Dev Biol, 2025, 12:1520949.
[89]
Singh A, Kumar A. Microglial inhibitory mechanism of coenzyme Q10 against abeta (1-42) induced cognitive dysfunctions: possible behavioral, biochemical, cellular, and histopathological alterations[J]. Front Pharmacol, 2015, 6:268.
[90]
Li W, Wang S zhang H, et al. Honokiol restores microglial phagocytosis by reversing metabolic reprogramming[J]. J Alzheimers Dis, 2021, 82(4):1475-1485.
[91]
Ma M, Wang J, Guo K, et al. A self-reinforced "microglia energy modulator" for synergistic amyloid-β clearance in Alzheimer's disease model[J]. Angew Chem Int Ed Endl, 2025, 137(8):1-9.
[92]
Cummings JL, Zhou Y, Lee G, et al. Alzheimer's disease drug development pipeline: 2025[J]. Alzheimers Dement (N Y), 2025, 11(2):e70098.
[93]
Stepanov YV, Golovynska I, Zhang R, et al. Near-infrared light reduces β-amyloid-stimulated microglial toxicity and enhances survival of neurons: mechanisms of light therapy for Alzheimer's disease[J]. Alzheimers Res Ther, 2022, 14(1):84.
[94]
NLi Q, Peng J, Luo Y, et al. Far infrared light irradiation enhances Aβ clearance via increased exocytotic microglial ATP and ameliorates cognitive deficit in Alzheimer's disease-like mice[J]. J Neuroinflammation, 2022, 19(1):145.
PDF(984 KB)

Accesses

Citation

Detail

Sections
Recommended

/