- Mini Review
- Open access
- Published:
Therapeutic implications of necroptosis activation in Alzheimer´s disease
Alzheimer's Research & Therapy volume 16, Article number: 275 (2024)
Abstract
In recent years, a growing body of research has unveiled the involvement of the necroptosis pathway in the pathogenesis of Alzheimer’s disease (AD). This evidence has shed light on the mechanisms underlying neuronal death in AD, positioning necroptosis at the forefront as a potential target for therapeutic intervention. This review provides an update on the current knowledge on this emerging, yet rapidly advancing topic, encompassing all published studies that present supporting proof of the role of the necroptosis pathway in the neurodegenerative processes of AD. The implication of misfolded tau and amyloid-β (Aβ) aggregates is highlighted, with evidence suggesting their direct or indirect involvement in necroptosis activation. In summary, the review underscores the significance of understanding the complex interplay between necroptosis, protein aggregates, and neurodegeneration in AD. The findings advocate for a comprehensive approach, combining therapeutic and early diagnostic strategies, to intervene in the disease process before irreversible damage occurs.
Background
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder and the leading cause of dementia [1]. Early onset (< 65 years) familial AD is caused by genetic mutations; however, this form of the disease is rare, accounting for less than 5% of cases [2,3,4,5]. The most common form is sporadic late-onset (> 65 years) AD, the cause of which remains unknown, although it has been linked to several risk factors. The greatest risk factors for sporadic AD include older age, genetics (several genes have been identified as increasing the risk of AD, with the ε4 allele of the apolipoprotein E gene having the most significant impact), and having a family history of AD [6,7,8,9]. Moreover, several modifiable risk factors have been linked to the disease, including obesity, physical inactivity, and low education, among others [10].
AD is characterized by progressive cognitive deterioration involving severe memory decline. Brain accumulation of senile plaques composed of aggregated amyloid-β (Aβ), and neurofibrillary tangles constituted of hyperphosphorylated tau, represents the neuropathological hallmark of the disease [11,12,13]. Prominent histopathological features of AD brains include neuroinflammation, synaptic loss, neurodegeneration, and neuronal death [11, 14,15,16,17,18], with the earliest alterations observed in the entorhinal cortex and hippocampus, followed by spread to various cortical areas [11, 19, 20]. Progressive ventricular enlargement with generalized cerebral atrophy can be observed in the AD brain as the result of massive neuronal loss [21]. Although the pathways underlying neuronal dysfunction and loss in AD remain elusive, emerging evidence has implicated the activation of several forms of programmed cell death in this process, including apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis [22,23,24,25,26]. Comprehensive studies are needed to determine how these cell death mechanisms interact to contribute to neuronal loss in AD and to identify whether distinct types of cell death are associated with different stages of disease progression. In this context, we specifically examine the role of the necroptosis pathway in neuronal loss in AD.
Necroptosis is a form of programmed cell death that occurs in response to cellular stressors, such as infection or tissue damage [27,28,29]. It is initiated by the activation of receptors such as tumor necrosis factor receptor 1 (TNFR1) or toll-like receptors (TLRs), leading to the formation of a signaling complex called the necrosome [30]. The necrosome consists of receptor-interacting protein kinase 1 (RIPK1) and RIPK3, which recruit and phosphorylate mixed lineage kinase domain-like protein (MLKL) [31]. Phosphorylated MLKL translocates to the plasma membrane, where it oligomerizes and disrupts membrane integrity, triggering inflammation and immune responses [32, 33]. In the last decade, there has been mounting evidence implicating the necroptosis pathway in the pathogenesis of various neurodegenerative diseases [34,35,36,37]. This review outlines the accumulating evidence suggesting necroptosis as a promising therapeutic avenue for AD.
Necroptosis as a potential therapeutic target for AD
The first proof of a role for necroptosis in AD came from a study that found an increase in the components of the necroptosis machinery in AD postmortem brain samples [37]. In addition, using the 5XFAD model of AD, reduced neuronal loss in mice treated with a necroptosis inhibitor was demonstrated [37]. The observation of necroptosis markers in AD brains was later confirmed by other research groups [38,39,40,41,42]. Notably, increasing studies have evidenced the functional implications of targeting necroptosis in experimental AD models, where blocking different components of the pathway (i.e. RIPK1, RIPK3, MLKL), by pharmacological or genetic means, has led to improvements in cognitive performance [41, 43,44,45]. Recent efforts to uncover the initiating factors triggering necroptosis in AD have suggested the involvement of TNF signaling. In this line, the upregulation of key molecules within the TNF pathway, including TNFR1, TNFR2, TNF and FADD, was observed in AD brains, and interestingly, the increased TNFR1 expression occurred specifically in pMLKL and pRIPK3-bearing neurons [38]. Accordingly, intracerebral administration of TNF-α induced necroptosis activation and neuronal loss in the hippocampus of wild-type mice and exacerbated these findings in the APP/PS1 model of AD [40].
While most studies on necroptosis activation in AD have predominantly identified its induction in neurons [37, 41, 42], emerging evidence suggests a broader involvement of the pathway in other cell types. In this context, a study demonstrated increased MLKL levels mostly in microglia following intrahippocampal injection of Aβ oligomers in APP23 mice [46]. Another work provided evidence for the expression of RIPK1, RIPK3 and MLKL in cultured human monocytes treated with Aβ aggregates [47]. In a recent study, necroptosis activation in cerebral endothelial cells of two transgenic AD models (APP/PS1 and APP-KINL−G−F) as well as in human AD brains was demonstrated, which was linked to reduced expression of mNat1/Nat2, a regulator of insulin sensitivity. Recovering cerebral mNAT1 inhibited endothelial necroptosis and improved cognitive function in APP-PS1 mice. In APP-KINL−G−F mice, pharmacological inhibition of RIPK1 reduced endothelial cell loss and cerebrovascular pathology [48]. Together, these studies shed light on contributions from non-neuronal cells to the pathogenic processes associated with AD. Thus, it is imperative to extend our scrutiny beyond neurons and explore the comprehensive landscape of necroptosis involvement across various cell populations, to unveil the potential multifaceted roles of this regulated necrotic pathway in the pathophysiology of AD.
Multiple cellular and molecular processes are altered in AD. Inflammation has been identified as a key factor driving neural injury and the progression of neurodegeneration. Glial cells play a crucial role in maintaining neuronal homeostasis, and their necroptosis-mediated dysfunction may exacerbate neuroinflammatory responses, leading to neuronal injury [49, 50]. Specifically, necroptosis in microglia may promote the release of pro-inflammatory cytokines, which could enhance synaptic loss and neuronal death through the activation of neurotoxic pathways. As mentioned above, necroptosis has been shown to contribute to cerebrovascular damage [48], which is also a significant contributor to neurodegeneration [51]. To fully comprehend the therapeutic potential of inhibiting necroptosis in AD, it is essential to conduct studies that integrate the various aspects of necroptotic signaling in both neuronal and non-neuronal cells. This will allow for a more comprehensive evaluation of how necroptosis contributes to neurodegeneration. In this context, inhibiting the necroptosis pathway could represent a multi-cellular therapeutic strategy, offering neuroprotection either directly by modulating necroptosis-mediated neuronal death or indirectly by targeting non-neuronal cells affected by necroptosis that contribute to neurodegenerative processes.
Most of the studies focusing on the role of necroptosis in AD have consistently demonstrated either direct or indirect involvement of protein aggregates in necroptosis activation. Histological analyses of AD brains have shown high colocalization between tau tangles and necroptotic markers [37, 42]. Moreover, the expression of hyperphosphorylated tau promoted the formation of the necrosome in cell cultures [52]. Additionally, necroptosis activation was demonstrated in the brains of TauP301S mice, which express mutant human tau and develop widespread neurofibrillary tangle-like inclusions [52]. In both in vitro and in vivo models, necroptosis triggered the production of pro-inflammatory cytokines and chemokines including IL-6, IL-15, IFN-1, TNF-α, Ccl2, Ccl5, Cxcl9, and Cxcl10. Necroptosis inhibition reduced the overexpression of these neuroinflammation mediators, as well as pTau-induced neuronal death and microglia hyperactivation, which was assessed by western blotting, qPCR and flow cytometry [52]. Likewise, the burden of Aβ oligomers correlates with necroptosis markers in AD brain samples [41]. Moreover, the injection of Aβ oligomers activated necroptosis, leading to neurodegeneration and memory alterations in wild-type mice [41] and worsened these alterations in APP23 mice [46]. Additional evidence came from studies using transgenic animals based on brain Aβ accumulation. In this context, neuronal necroptosis has been demonstrated in the 5XFAD and APP21 mouse lines, which develop extensive Aβ pathology [37, 46]. In addition, Aβ-induced necroptosis has also been demonstrated in cultured neurons [53,54,55]. A step forward toward unveiling the mechanisms involved was recently achieved as it was demonstrated that Aβ deposition drives tau tangle formation and necroptosis-mediated neuronal death in human neurons xenografted in an APP knock-in mouse model of AD. Notably, blocking the maternally expressed gene 3 (MEG3) reduced the expression of necroptosis markers in the xenografted neurons. This data supports the view of Aβ deposition as the initial trigger of neuropathology in AD and provides proof that necroptosis is induced by MEG3 up-regulation, which occurs downstream of tau accumulation [56]. Collectively, these findings represent compelling evidence demonstrating that both Aβ and tau aggregates serve as catalysts for the initiation of the necroptotic pathway in AD (Fig. 1).
Although current preclinical data is compelling regarding the role of necroptosis as the primary mechanism by which neurons die in the context of AD, the translation of this evidence to the clinic is still in its initial phase. DNL747, developed by Denali Therapeutics, was the first RIPK1 inhibitor tested in a clinical trial for AD. This small molecule was well-tolerated in healthy volunteers and was then tested in two phase I trials involving 16 AD patients. While the drug was safe and well tolerated in both trials, the study indicated that a higher dose would be needed to achieve therapeutic efficacy, which could implicate a potential safety risk [57]. For this reason, it was announced that the studies will be shifted to test the successor molecule DNL788, another inhibitor of RIPK1 with a superior preclinical therapeutic window compared to DNL747. A recent report on the findings of a phase I trial performed on healthy participants revealed that the drug was generally safe and well-tolerated following single and repeated oral doses, supporting further development [58]. Similar results were obtained in a clinical study of SIR-2446, an additional RIPK1 inhibitor developed by Sironax Therapeutics for treating AD and multiple sclerosis, providing support for its continued clinical advancement [59].
Necroptosis activation in AD. Protein misfolding and accumulation leads to activation of the TNF signaling pathway. TNFR1 activation results in the recruitment of RIPK1. In the absence of caspase-8 activity, RIPK1 interacts with RIPK3 to form the necrosome. MLKL is recruited to the complex and phosphorylated by pRIPK3, which triggers its oligomerization and translocation to the plasma membrane, where executes necroptosis
While most necroptosis inhibitors currently in clinical trials for various diseases target RIPK1, several compounds targeting the downstream kinase RIPK3, including ponatinib, dabrafenib, and sorafenib, have been approved for cancer treatment [60,61,62]. Although these drugs are not specific RIPK3 inhibitors, preclinical data point to the importance of assessing their efficacy in AD patients [56, 60]. Similarly, although MLKL is the final effector in necroptosis and therefore an important target, there are currently no clinical trials investigating this in the context of AD.
One of the important lessons learned from numerous failed clinical trials for AD is that attempting to revert the extensive damage present in the late stages of the disease, in an effort to recover brain function, is unlikely [61, 62]. An effective treatment before the symptoms arise seems to be a more reasonable approach than attempting to reverse the pathology when the damage is irreversible due to the complexity of the pathological processes triggered [63]. Thus, the simultaneous development of therapeutic and early diagnosis strategies is crucial to efficiently tackle the disease while it is still possible to halt the neurodegeneration process. It remains to be established whether the necroptotic pathway is activated in the preclinical stages of AD and, if so, whether it could serve as an early biomarker or target for the disease. Yet, the evidence indicates a link between necroptosis and protein aggregation, which is largely recognized as an early or even initial event in AD pathogenesis [64]. Moreover, there is evidence that the necroptotic signaling mediates axonal degeneration [34, 65,66,67], and numerous studies suggest that axonal pathology precedes the clinical manifestation of AD [68,69,70,71,72,73]. However, the potential involvement of necroptosis in axonal degeneration has not been demonstrated in the context of AD.
Considering that multiple pathways are affected in AD, the need for combined therapies becomes evident. By targeting different mechanisms, such as protein aggregation and necroptosis, a combined treatment may have a higher chance of success in both improving symptoms and delaying the progression of neurodegeneration. Current evidence strongly supports a role for necroptosis in the pathology of AD, however, further studies are needed to determine the possible implication of the necroptosis pathway in the initial phases of neurodegeneration in the disease.
Conclusions
The evidence presented underscores the significant involvement of necroptosis in AD pathology. Studies have consistently shown the upregulation of necroptosis markers in AD brains and demonstrated the beneficial effects of inhibiting this pathway in experimental models, highlighting its potential as a therapeutic target. Despite promising preclinical findings, clinical translation of necroptosis-targeting therapies is still in the early stages, emphasizing the need for further research to elucidate the role of necroptosis in early neurodegenerative processes.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- AD:
-
Alzheimer’s disease
- MEG3:
-
maternally expressed gene 3
- TNFR1:
-
tumor necrosis factor receptor 1
- TLRs:
-
toll-like receptors
- RIPK1:
-
receptor-interacting protein kinase 1
- MLKL:
-
mixed lineage kinase domain-like protein
References
Alzheimer’s A. 2023 Alzheimer’s disease facts and figures. Alzheimers Dement [Internet]. 2023;19(4):1598–695. http://www.ncbi.nlm.nih.gov/pubmed/36918389
Citron M, Oltersdorf T, Haass C, McConlogue L, Hung AY, Seubert P et al. Mutation of the β-amyloid precursor protein in familial Alzheimer’s disease increases β-protein production. Nature [Internet]. 1992;360(6405):672–4. http://www.nature.com/articles/360672a0
Hendriks L, van Duijn CM, Cras P, Cruts M, Van Hul W, van Harskamp F, et al. Presenile dementia and cerebral haemorrhage linked to a mutation at codon 692 of the β–amyloid precursor protein gene. Nat Genet. 1992;1(3):218–21.
Suzuki N, Cheung T, Cai X, Odaka A, Otvos L, Eckman C et al. An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science (1979) [Internet]. 1994;264(5163):1336–40. http://www.nature.com/articles/nrd2896
Harvey RJ, Skelton-Robinson M, Rossor MN. The prevalence and causes of dementia in people under the age of 65 years. J Neurol Neurosurg Psychiatry. 2003;74(9):1206–9.
Farrer LA. Effects of Age, Sex, and Ethnicity on the Association Between Apolipoprotein E Genotype and Alzheimer Disease. JAMA [Internet]. 1997;278(16):1349. http://jama.jamanetwork.com/article.aspx?doi=10.1001/jama.1997.03550160069041
Haney MS, Pálovics R, Munson CN, Long C, Johansson PK, Yip O, et al. APOE4/4 is linked to damaging lipid droplets in Alzheimer’s disease microglia. Nature. 2024;628(8006):154–61.
Alzheimer A. 2024 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia [Internet]. 2024;20(5):3708–821. https://alz-journals.onlinelibrary.wiley.com/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1002/alz.13809
Lautenschlager NT, Cupples LA, Rao VS, Auerbach SA, Becker R, Burke J et al. Risk of dementia among relatives of Alzheimer’s disease patients in the MIRAGE study. Neurology [Internet]. 1996;46(3):641–50. https://www.neurology.org/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1212/WNL.46.3.641
Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. The Lancet. Volume 396. Lancet Publishing Group; 2020. pp. 413–46.
Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in alzheimer disease: Age categories from 1 to 100 years. J Neuropathol Exp Neurol. 2011;70(11):960–9.
Jack CR, Knopman DS, Jagust WJ, Petersen RC, Weiner MW, Aisen PS et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. 12, The Lancet Neurology. 2013. pp. 207–16.
Gordon BA, Blazey TM, Su Y, Hari-Raj A, Dincer A, Flores S, et al. Spatial patterns of neuroimaging biomarker change in individuals from families with autosomal dominant Alzheimer’s disease: a longitudinal study. Lancet Neurol. 2018;17(3):241–50.
Heneka MT, Carson MJ, Khoury J, El, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405.
Spires-Jones TL, Hyman BT. The Intersection of Amyloid Beta and Tau at Synapses in Alzheimer’s Disease. Neuron [Internet]. 2014;82(4):756–71. https://linkinghub.elsevier.com/retrieve/pii/S0896627314003900
Gouras GK, Tampellini D, Takahashi RH, Capetillo-Zarate E. Intraneuronal β-amyloid accumulation and synapse pathology in Alzheimer’s disease. Acta Neuropathol [Internet]. 2010;119(5):523–41. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3624763/pdf/nihms412728.pdf
DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol. 1990;27(5):457–64.
Collins-Praino LE, Francis YI, Griffith EY, Wiegman AF, Urbach J, Lawton A, et al. Soluble amyloid beta levels are elevated in the white matter of Alzheimer’s patients, independent of cortical plaque severity. Acta Neuropathol Commun. 2014;2(1):1–10.
Andrade-Moraes CH, Oliveira-Pinto AV, Castro-Fonseca E, da Silva CG, Guimarães DM, Szczupak D, et al. Cell number changes in Alzheimer’s disease relate to dementia, not to plaques and tangles. Brain. 2013;136(Pt 12):3738–52.
Thal DR, Rüb U, Orantes M, Braak H. Phases of Aβ-deposition in the human brain and its relevance for the development of AD. Neurology [Internet]. 2002;58(12):1791–800. http://www.neurology.org/lookup/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1212/WNL.58.12.1791
Blinkouskaya Y, Weickenmeier J. Brain shape changes Associated with cerebral atrophy in healthy aging and Alzheimer’s Disease. Front Mech Eng. 2021;7.
Moujalled D, Strasser A, Liddell JR. Molecular mechanisms of cell death in neurological diseases. Cell Death and Differentiation. Volume 28. Springer Nature; 2021. pp. 2029–44.
Brokaw DL, Piras IS, Mastroeni D, Weisenberger DJ, Nolz J, Delvaux E, et al. Cell death and survival pathways in Alzheimer’s disease: an integrative hypothesis testing approach utilizing -omic data sets. Neurobiol Aging. 2020;95:15–25.
Moonen S, Koper MJ, Van Schoor E, Schaeverbeke JM, Vandenberghe R, von Arnim CAF, et al. Pyroptosis in Alzheimer’s disease: cell type-specific activation in microglia, astrocytes and neurons. Acta Neuropathol. 2023;145(2):175–95.
Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013;19:983–97.
Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, et al. Ferroptosis and its role in Diverse Brain diseases. Mol Neurobiol. 2019;56(7):4880–93.
Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M et al. Phosphorylation-Driven Assembly of the RIP1-RIP3 Complex Regulates Programmed Necrosis and Virus-Induced Inflammation. Cell [Internet]. 2009;137(6):1112–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2009.05.037
Degterev A, Hitomi J, Germscheid M, Ch’ IL, Korkina O, Teng X et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol [Internet]. 2008;4(5):313–21. http://www.nature.com/doifinder/https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nchembio.83
Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol [Internet]. 2005;1(2):112–9. http://www.nature.com/doifinder/https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nchembio711
He S, Wang L, Miao L, Wang T, Du F, Zhao L, et al. Receptor interacting protein Kinase-3 determines Cellular necrotic response to TNF-α. Cell. 2009;137(6):1100–11.
Rodriguez DA, Weinlich R, Brown S, Guy C, Fitzgerald P, Dillon CP et al. Characterization of RIPK3-mediated phosphorylation of the activation loop of MLKL during necroptosis. Cell Death Differ [Internet]. 2016;23(1):76–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cdd.2015.70
Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J et al. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol [Internet]. 2013;16(1):55–65. http://www.nature.com/doifinder/https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncb2883
Dondelinger Y, Declercq W, Montessuit S, Roelandt R, Goncalves A, Bruggeman I et al. MLKL Compromises Plasma Membrane Integrity by Binding to Phosphatidylinositol Phosphates. Cell Rep [Internet]. 2014;7(4):971–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2014.04.026
Oñate M, Catenaccio A, Salvadores N, Saquel C, Martinez A, Moreno-Gonzalez I et al. The necroptosis machinery mediates axonal degeneration in a model of Parkinson disease. Cell Death Differ [Internet]. 2019; https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41418-019-0408-4
Ofengeim D, Ito Y, Najafov A, Zhang Y, Shan B, DeWitt JP, et al. Activation of necroptosis in multiple sclerosis. Cell Rep. 2015;10(11):1836–49.
Ito Y, Ofengeim D, Najafov A, Das S, Saberi S, Li Y et al. RIPK1 mediates axonal degeneration by promoting inflammation and necroptosis in ALS. Science (1979) [Internet]. 2016;353(6299):603–8. http://www.sciencemag.org/lookup/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.aaf6803
Caccamo A, Branca C, Piras IS, Ferreira E, Huentelman MJ, Liang WS, et al. Necroptosis activation in Alzheimer’s disease. Nat Neurosci. 2017;20(9):1236–46.
Jayaraman A, Htike TT, James R, Picon C, Reynolds R. TNF-mediated neuroinflammation is linked to neuronal necroptosis in Alzheimer’s disease hippocampus. Acta Neuropathol Commun. 2021;9(1).
Koper MJ, Tomé SO, Gawor K, Belet A, Van Schoor E, Schaeverbeke J et al. LATE-NC aggravates GVD-mediated necroptosis in Alzheimer’s disease. Acta Neuropathol Commun. 2022;10(1).
Xu C, Wu J, Wu Y, Ren Z, Yao Y, Chen G, et al. TNF-α-dependent neuronal necroptosis regulated in Alzheimer’s disease by coordination of RIPK1-p62 complex with autophagic UVRAG. Theranostics. 2021;11(19):9452–69.
Salvadores N, Moreno-Gonzalez I, Gamez N, Quiroz G, Vegas-Gomez L, Escandón M et al. Aβ oligomers trigger necroptosis-mediated neurodegeneration via microglia activation in Alzheimer’s disease. Acta Neuropathol Commun. 2022;10(1).
Koper MJ, Van Schoor E, Ospitalieri S, Vandenberghe R, Vandenbulcke M, von Arnim CAF, et al. Necrosome complex detected in granulovacuolar degeneration is associated with neuronal loss in Alzheimer’s disease. Acta Neuropathol. 2020;139(3):463–84.
Motawi TMK, Abdel-Nasser ZM, Shahin NN. Ameliorative effect of Necrosulfonamide in a rat model of Alzheimer’s Disease: targeting mixed lineage kinase domain-like protein-mediated necroptosis. ACS Chem Neurosci. 2020;11(20):3386–97.
Abdelhady R, Younis NS, Ali O, Shehata S, Sayed RH, Nadeem RI. Cognitive enhancing effects of pazopanib in Dgalactose/ovariectomized Alzheimer’s rat model: insights into the role of RIPK1/RIPK3/MLKL necroptosis signaling pathway. Inflammopharmacology. 2023;31(5):2719–29.
Sun Y, Xu L, Shao H, Quan D, Mo Z, Wang J, et al. Discovery of a Trifluoromethoxy Cyclopentanone Benzothiazole receptor-interacting protein kinase 1 inhibitor as the treatment for Alzheimer’s Disease. J Med Chem. 2022;65(21):14957–69.
Yu H, Morihara R, Ota-Elliott R, Bian Z, Bian Y, Hu X et al. Injection of exogenous amyloid-β oligomers aggravated cognitive deficits, and activated necroptosis, in APP23 transgenic mice. Brain Res. 2023;1821.
Yuan S, Li H, Yang C, Xie W, Wang Y, Zhang J et al. DHA attenuates Aβ-induced necroptosis through the RIPK1/RIPK3 signaling pathway in THP-1 monocytes. Biomed Pharmacotherapy. 2020;126.
Zou C, Mifflin L, Hu Z, Zhang T, Shan B, Wang H et al. Reduction of mNAT1/hNAT2 contributes to cerebral endothelial necroptosis and Aβ Accumulation in Alzheimer’s Disease. Cell Rep. 2020;33(10).
Ofengeim D, Mazzitelli S, Ito Y, DeWitt JP, Mifflin L, Zou C, et al. RIPK1 mediates a disease-associated microglial response in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2017;114(41):E8788–97.
Huang Z, Zhou T, Sun X, Zheng Y, Cheng B, Li M et al. Necroptosis in microglia contributes to neuroinflammation and retinal degeneration through TLR4 activation. Cell Death Differ [Internet]. 2018;25(1):180–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cdd.2017.141
Yew B, Nation DA. Cerebrovascular resistance: effects on cognitive decline, cortical atrophy, and progression to dementia. http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf
Dong Y, Yu H, Li X, Bian K, Zheng Y, Dai M et al. Hyperphosphorylated tau mediates neuronal death by inducing necroptosis and inflammation in Alzheimer’s disease. J Neuroinflammation. 2022;19(1).
Liu K, Chojnacki JE, Wade EE, Saathoff JM, Lesnefsky EJ, Chen Q, et al. Bivalent compound 17MN exerts neuroprotection through interaction at multiple sites in a cellular model of Alzheimer’s disease. J Alzheimer’s Disease. 2015;47(4):1021–33.
Wang W, Gu X, Cheng Z, Lu X, Xie S, Liu X. IKKβ Alleviates Neuron Injury in Alzheimer’s Disease via regulating autophagy and RIPK1-Mediated necroptosis. Mol Neurobiol. 2022;59(4):2407–23.
Li XX, Lang XY, Ren TT, Wang J, Lan R, Qin XY. Coeloglossum viride var. Bracteatum extract attenuates Aβ-induced toxicity by inhibiting RIP1–driven inflammation and necroptosis. J Ethnopharmacol. 2022;282.
Balusu S, Horré K, Thrupp N, Craessaerts K, Snellinx A, Serneels L, et al. MEG3 activates necroptosis in human neuron xenografts modeling Alzheimer’s disease. Sci (1979). 2023;381(6663):1176–82.
Vissers MFJM, Heuberger JAAC, Groeneveld GJ, Oude Nijhuis J, De Deyn PP, Hadi S, et al. Safety, pharmacokinetics and target engagement of novel RIPK1 inhibitor SAR443060 (DNL747) for neurodegenerative disorders: Randomized, placebo-controlled, double-blind phase I/Ib studies in healthy subjects and patients. Clin Transl Sci. 2022;15(8):2010–23.
Hincelin-Mery A, Nicolas X, Cantalloube C, Pomponio R, Lewanczyk P, Benamor M et al. Safety, pharmacokinetics, and target engagement of a brain penetrant RIPK1 inhibitor, SAR443820 (DNL788), in healthy adult participants. Clin Transl Sci. 2023.
Sun ALA, Gillies JD, Shen Y, Deng H, Xue F, Ma Y et al. A phase I randomized study to evaluate safety, pharmacokinetics, and pharmacodynamics of SIR2446M, a selective RIPK1 inhibitor, in healthy participants. Clin Transl Sci. 2024;17(7).
Koper MJ, Moonen S, Ronisz A, Ospitalieri S, Callaerts-Vegh Z, T’Syen D et al. Inhibition of an Alzheimer’s disease–associated form of necroptosis rescues neuronal death in mouse models. Sci Transl Med. 2024;16(771).
Mehta D, Jackson R, Paul G, Shi J, Sabbagh M. Why do trials for Alzheimer’s disease drugs keep failing? A discontinued drug perspective for 2010–2015. Expert Opin Investig Drugs. 2017;26(6):735–9.
Yiannopoulou KG, Anastasiou AI, Zachariou V, Pelidou SH. Reasons for failed trials of disease-modifying treatments for alzheimer disease and their contribution in recent research. Volume 7. Biomedicines. MDPI AG; 2019.
Cummings J, Aisen P, Apostolova LG, Atri A, Salloway S, Weiner M. Aducanumab: appropriate use recommendations. J Prev Alzheimer’s Disease. 2021;8(4):398–410.
Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med [Internet]. 2016;8(6):595–608. http://embomolmed.embopress.org/lookup/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.15252/emmm.201606210
Hernández DE, Salvadores NA, Moya-Alvarado G, Catalán RJ, Bronfman FC, Court FA. Axonal degeneration induced by glutamate excitotoxicity is mediated by necroptosis. J Cell Sci [Internet]. 2018;131(22):jcs214684. http://jcs.biologists.org/lookup/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.214684
Arrázola MS, Saquel C, Catalán RJ, Barrientos SA, Hernandez DE, Martínez NW et al. Axonal Degeneration Is Mediated by Necroptosis Activation. The Journal of Neuroscience [Internet]. 2019;39(20):3832–44. http://www.jneurosci.org/lookup/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.0881-18.2019
Ko KW, Milbrandt J, DiAntonio A. SARM1 acts downstream of neuroinflammatory and necroptotic signaling to induce axon degeneration. Journal of Cell Biology [Internet]. 2020;219(8). https://rupress.org/jcb/article/219/8/e201912047/151915/SARM1-acts-downstream-of-neuroinflammatory-and
Stokin GB. Axonopathy and Transport Deficits Early in the Pathogenesis of Alzheimer’s Disease. Science (1979) [Internet]. 2005;307(5713):1282–8. http://www.sciencemag.org/cgi/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.1105681
Cras P, Kawai M, Lowery D, Gonzalez-DeWhitt P, Greenberg B, Perry G. Senile plaque neurites in Alzheimer disease accumulate amyloid precursor protein. Proceedings of the National Academy of Sciences [Internet]. 1991;88(17):7552–6. http://www.pnas.org/cgi/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.88.17.7552
Dessi F, Colle MA, Hauw JJ, Duyckaerts C. Accumulation of SNAP-25 immunoreactive material in axons of Alzheimer’s disease. NeuroReport. 1997;8(17):3685–9.
Stoub TR, DeToledo-Morrell L, Stebbins GT, Leurgans S, Bennett DA, Shah RC. Hippocampal disconnection contributes to memory dysfunction in individuals at risk for Alzheimer’s disease. Proceedings of the National Academy of Sciences [Internet]. 2006;103(26):10041–5. http://www.pnas.org/cgi/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0603414103
Rogalski EJ, Murphy CM, Detoledo-Morrell L, Shah RC, Moseley ME, Bammer R, et al. Changes in parahippocampal white matter integrity in amnestic mild cognitive impairment: a diffusion tensor imaging study. Behav Neurol. 2009;21(1–2):51–61.
Kalus P, Slotboom J, Gallinat J, Mahlberg R, Cattapan-Ludewig K, Wiest R, et al. Examining the gateway to the limbic system with diffusion tensor imaging: the perforant pathway in dementia. NeuroImage. 2006;30(3):713–20.
Acknowledgements
Not applicable.
Funding
No funding was received towards this work.
Author information
Authors and Affiliations
Contributions
N.S. conceptually designed the work. E.C. and N.S. wrote the manuscript. E.C. made the illustration. N.S. edited the manuscript and figure. Both authors read and approved the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Carrazana, E., Salvadores, N. Therapeutic implications of necroptosis activation in Alzheimer´s disease. Alz Res Therapy 16, 275 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13195-024-01649-8
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13195-024-01649-8