4,772
Views
10
CrossRef citations to date
0
Altmetric
Toxicology

Role of neurotoxicants in the pathogenesis of Alzheimer’s disease: a mechanistic insight

, , , , , , & ORCID Icon show all
Pages 1479-1504 | Received 25 May 2021, Accepted 04 Aug 2021, Published online: 25 Aug 2021

Figures & data

Figure 1. Aβ-mediated neurotoxicity in Alzheimer’s disease. The major neurotoxic effects of Aβ1-42 peptides in the pathological development of AD are illustrated in this diagram. That includes the formation of soluble Aβ oligomers, amyloid plaques and neurofibrillary tangles, generation of reactive oxygen species, the imbalanced concentration of metal ions, and functional impairment of mitochondria, brain immune cells, and neurotransmission. Through these mechanisms, toxic Aβ induces oxidative stress, death of neurons, synaptic malfunction, neuroinflammation, and exacerbates the abnormal protein aggregation in critical brain regions.

Figure 1. Aβ-mediated neurotoxicity in Alzheimer’s disease. The major neurotoxic effects of Aβ1-42 peptides in the pathological development of AD are illustrated in this diagram. That includes the formation of soluble Aβ oligomers, amyloid plaques and neurofibrillary tangles, generation of reactive oxygen species, the imbalanced concentration of metal ions, and functional impairment of mitochondria, brain immune cells, and neurotransmission. Through these mechanisms, toxic Aβ induces oxidative stress, death of neurons, synaptic malfunction, neuroinflammation, and exacerbates the abnormal protein aggregation in critical brain regions.

Figure 2. Major neurotoxicants at the onset and progression of Alzheimer’s disease. The abnormal build-up of amyloid-beta protein that generates amyloid plaques and hyperphosphorylated tau protein that forms neurofibrillary tangles in and around neurons are apparent at the onset of Alzheimer’s disease. Multiple neurotoxicants such as metals, pesticides, and nanoparticles have been found to augment the formation of Aβ aggregates and NFTs through different mechanisms. These neurotoxicants produce oxidative stress in neurons that trigger Aβ peptide formation and hyperphosphorylate tau protein. Neurotoxicants stabilise APP expression and β- secretase enzyme activities; on the other hand, they disrupt the functions of antioxidant enzymes, Aβ degrading proteins, and receptors that result in amyloid plaque formation. Neurotoxicants bind with tau, dissociate them from microtubules and increase their hyperphosphorylation. Enzymes that dephosphorylate tau protein are also inhibited by neurotoxicants that ultimately leads to the formation of neurofibrillary tangles.

Figure 2. Major neurotoxicants at the onset and progression of Alzheimer’s disease. The abnormal build-up of amyloid-beta protein that generates amyloid plaques and hyperphosphorylated tau protein that forms neurofibrillary tangles in and around neurons are apparent at the onset of Alzheimer’s disease. Multiple neurotoxicants such as metals, pesticides, and nanoparticles have been found to augment the formation of Aβ aggregates and NFTs through different mechanisms. These neurotoxicants produce oxidative stress in neurons that trigger Aβ peptide formation and hyperphosphorylate tau protein. Neurotoxicants stabilise APP expression and β- secretase enzyme activities; on the other hand, they disrupt the functions of antioxidant enzymes, Aβ degrading proteins, and receptors that result in amyloid plaque formation. Neurotoxicants bind with tau, dissociate them from microtubules and increase their hyperphosphorylation. Enzymes that dephosphorylate tau protein are also inhibited by neurotoxicants that ultimately leads to the formation of neurofibrillary tangles.

Table 1. Signalling pathways modulated by neurotoxicants in the pathogenesis of Alzheimer’s disease.

Figure 3. Inhibition of the Wnt/β-catenin signalling by neurotoxicants. In normal brain, when Wnt signalling is switched on, GSK-3β is found to be inactive, and tau protein remains dephosphorylated. β-catenin translocation in the nucleus activates Wnt target genes that inhibit the development of Aβ1–42. In the presence of various neurotoxicants (Fe, Cu, As, Pb, MeHg, BPA, pesticides, and NPs), Dkk1 and GSK-3β, the inhibitors of Wnt signalling cascade become activated. Activation of Wnt proteins, LRP 5/6, and β-catenin are also inhibited by some neurotoxicants (BPA, Sb). β-catenin is phosphorylated by GSK-3β and undergoes proteasomal degradation. As a result, Wnt signalling is shut off, leading to tau hyperphosphorylation and Aβ1–42 production and aggregation that aids AD pathology.

Figure 3. Inhibition of the Wnt/β-catenin signalling by neurotoxicants. In normal brain, when Wnt signalling is switched on, GSK-3β is found to be inactive, and tau protein remains dephosphorylated. β-catenin translocation in the nucleus activates Wnt target genes that inhibit the development of Aβ1–42. In the presence of various neurotoxicants (Fe, Cu, As, Pb, MeHg, BPA, pesticides, and NPs), Dkk1 and GSK-3β, the inhibitors of Wnt signalling cascade become activated. Activation of Wnt proteins, LRP 5/6, and β-catenin are also inhibited by some neurotoxicants (BPA, Sb). β-catenin is phosphorylated by GSK-3β and undergoes proteasomal degradation. As a result, Wnt signalling is shut off, leading to tau hyperphosphorylation and Aβ1–42 production and aggregation that aids AD pathology.

Figure 4. Dysregulation of autophagy and mTOR signalling by neurotoxicants. Although autophagy and mTOR signalling are vital for the healthy and normal functioning of neurons, some neurotoxicants interrupt their regulation. Excessive activation of mTOR signalling through neurotoxicants (Mn, Cd) results in mitophagy and neuronal apoptosis that inhibits normal autophagic function. However, autophagy-related proteins, including ATG, Beclin, LC3II, etc. and different autophagic steps are negatively influenced by neurotoxicants (Fe, Cu, Mn, As, Pb, MeHg), leading to the uncontrolled autophagic influx. Some neurotoxicants (As, MeHg, Mn) damage lysosomal structure, which after fusion with autophagosome, produces immature autophagolysosome vacuoles (AVs) and halt the degradation of autophagolysosomes. Finally, increased accumulation of AVs in neurons triggers neuronal death.

Figure 4. Dysregulation of autophagy and mTOR signalling by neurotoxicants. Although autophagy and mTOR signalling are vital for the healthy and normal functioning of neurons, some neurotoxicants interrupt their regulation. Excessive activation of mTOR signalling through neurotoxicants (Mn, Cd) results in mitophagy and neuronal apoptosis that inhibits normal autophagic function. However, autophagy-related proteins, including ATG, Beclin, LC3II, etc. and different autophagic steps are negatively influenced by neurotoxicants (Fe, Cu, Mn, As, Pb, MeHg), leading to the uncontrolled autophagic influx. Some neurotoxicants (As, MeHg, Mn) damage lysosomal structure, which after fusion with autophagosome, produces immature autophagolysosome vacuoles (AVs) and halt the degradation of autophagolysosomes. Finally, increased accumulation of AVs in neurons triggers neuronal death.

Figure 5. Alterations in PKC signalling by neurotoxicants. PKC signalling is associated with multiple functions, including the formation of sAPPα and inhibition of GSK-3β that decreases Aβ production and tau hyperphosphorylation. At the same time, some neurotoxic agents (Al, Pb, Ni, Hg, MeHg) interfere with PKC enzyme expressions and activities that alter normal PKC signalling. This, in turn, enhances Aβ1–42 production and directly affects PKC signalling.

Figure 5. Alterations in PKC signalling by neurotoxicants. PKC signalling is associated with multiple functions, including the formation of sAPPα and inhibition of GSK-3β that decreases Aβ production and tau hyperphosphorylation. At the same time, some neurotoxic agents (Al, Pb, Ni, Hg, MeHg) interfere with PKC enzyme expressions and activities that alter normal PKC signalling. This, in turn, enhances Aβ1–42 production and directly affects PKC signalling.

Table 2. Role of neurotoxin-induced cellular stresses in the progression of Alzheimer’s disease.

Table 3. Effects of neurotoxic agents on iGluRs and mGluRs in the onset and progression of Alzheimer’s disease.

Data availability statement

Data available on request from the authors.