Skip to main content
Erschienen in: Translational Neurodegeneration 1/2018

Open Access 01.12.2018 | Review

Alzheimer’s disease hypothesis and related therapies

verfasst von: Xiaoguang Du, Xinyi Wang, Meiyu Geng

Erschienen in: Translational Neurodegeneration | Ausgabe 1/2018

Abstract

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause for dementia. There are many hypotheses about AD, including abnormal deposit of amyloid β (Aβ) protein in the extracellular spaces of neurons, formation of twisted fibers of tau proteins inside neurons, cholinergic neuron damage, inflammation, oxidative stress, etc., and many anti-AD drugs based on these hypotheses have been developed. In this review, we will discuss the existing and emerging hypothesis and related therapies.
Abkürzungen
AD
Alzheimer’s disease
Amyloid β
CNS
Central nervous system
CSF1R
Colony stimulating factor 1 receptor
GABA
Gamma-aminobutyric acid
IVIG
Intravenous immunoglobulin
MAMP
Microbes associated molecular patterns
NFTs
Neurofibrillary tangles
NMDA
N-methyl-D-aspartate
NSAIDs
Non-steroid anti-inflammatory drugs
O-GlcNAc
O-linked N-acetylglucosamine
SCFAs
Short-chain-fatty acids
Th17
T helper lymphocytes 17

Background

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder, which is the most common cause for dementia and imposes immense suffering on patients and their families. According to the World Alzheimer Report 2016, there are currently about 46.8 million people suffering with AD worldwide. The ageing of world population will further compound this problem and lead to a steep increase in the number of AD patients. The numbers of AD patients are expected to double nearly every 20 years, and thereby the population of AD will reach 74.7 million in 2030 and 131.5 million in 2050 [1]. AD has become the third major cause of disability and death for the elderly, only after cardiovascular and cerebrovascular diseases and malignant tumors.
However, only five drugs have been approved by the FDA to treat AD over the past hundred years since the first AD patient was diagnosed. Not only that, these approved drugs including cholinesterase inhibitors, N-methyl-D-aspartate (NMDA) receptor antagonist or their combination usually provide temporary and incomplete symptomatic relief accompanied with severe side effects. The marginal benefits were unable to slow the progression of AD. Thus, developing drugs for more effective AD treatment is in urgent need.

Current hypothesis about AD and anti-AD drug development

AD is a complicated disease involving many factors. Due to the complexity of human brains, the lack of reasonable animal models and research tools, the detailed pathogenesis of AD is still unclear so far. Many hypotheses about AD have been developed, including amyloid β (Aβ), Tau, cholinergic neuron damage and oxidative stress, inflammation, etc. Thus, many efforts have been done to develop anti-AD drugs based on these hypotheses.

Aβ cascade hypothesis

Extracellular deposits of Aβ peptides as senile plaques, intraneuronal neurofibrillary tangles (NFTs), and large-scale neuronal loss were the main pathological features of AD. Thus, Aβ peptides have long been viewed as a potential target for AD which dominated new drug research during the past twenty years [2]. The most direct strategy in anti-Aβ therapy is to reduce Aβ production by targeting β- and γ-secretase [3]. Safety issues are the overriding problem. For targeting γ-secretase, undesirable side effects are inevitable due to its physiological substrates, eg. the Notch signaling protein [47], which is essential in normal biological process. Similarily, targeting β-secretase is also challenged for the side effects such as blindness and the large catalytic pocket [8]. More importantly, in sporadic AD cases, the majority of AD patients do not necessarily have over-producted amyloid precursor protein. Besides, Aβ isoforms could also serve as endogenous positive regulators for neurotransmitter release at hippocampal synapses [9]. Thus, inhibiting Aβ production may encounter many challenges.
Aβ clearance by immunotherapy is the alternative choice. For active Aβ-immunotherapy, although the first active AD vaccine (AN1792) developed by ELAN showed some beneficial effects such as less cognitive decline, it was suspended owing to serious side effect, or meningoencephalitis [1012]. Also, the passive immunotherapy did not do much better than active immunotherapy. Several antibodies targeting Aβ have failed in clinical trials, including bapineuzumab (Pfizer/Johnson & Johnson) [13, 14], Crenezumab (Genentech) [15, 16], solanezumab (Eli Lilly) [1618] and ponezumab (Johnson & Johnson /Pfizer) [1921]. In addition, although passive immunotherapy could overcome some problems of active immunotherapy, there were still inevitable side effects such as amyloid-related imaging abnormalities [22]. Likewise, the small molecule Aβ binder scyllo-inositol [23] and tramiprosate [2426] also failed in clinical trials. These failures even cast more doubts on the Aβ theory [27]. Actually, the strategy of targeting only a single functional subregion of Aβ may partly account for these failures [27, 28]. Furthermore, immunotherapy may influence the human immune system, which might cause beneficial or detrimental consequence (such as side effects). However, every cloud has a silver lining. A phase Ib trial of aducanumab (Biogen) showed a positive correlation between brain Aβ levels and disease exacerbation as measured by Clinical Dementia Rating [2931]. Even the failed phase III EXPEDITION3 trial of solanezumab (Eli Lilly) still demonstrated better performance in Clinical Dementia Rating Sum of Boxes and beneficial impacts on Mini-Mental State Examination and Activities of Daily Living [17, 18, 32, 33]. Thus, despite all kinds of problems, immunotherapy may still be the better approach to modify the extent of neurodegeneration in AD currently [34].
In fact, the original amyloid cascade hypothesis was that “Aβ is the causative agent in Alzheimer’s Disease pathology, and that neurofibrillary tangles, cell loss, vascular damage, and dementia follow as a direct result of this deposition” [35]. After decades of research, although the bulk of data still supports a role for Aβ as the primary initiator of the complex pathogenic cascade in AD, more and more evidences indicate that Aβ acts as a trigger in the early disease process and appears to be necessary but not sufficient in the late stage of AD [36]. Especially, recent rapid progresses in understanding on toxic amyloid assembly and Aβ metabolism associated systemic abnormalities will provide fresh impetus and new opportunities for this interesting approach [37].

Tau hypothesis

Neurofibrillary tangles, another intracellular hallmark of AD, are composed of tau. Tau is a microtubule-associated protein working as scaffolding proteins that are enriched in axons. In pathological conditions, tau aggregation will impair axons of neurons and thus cause neurodegeneration. After numerous failures of Aβ-targeting drugs for AD, more interests are turning to explore the therapeutic potential of targeting tau, particularly as studies of biomarkers suggest that tau pathology is more closely linked to the progression of AD [38].
Tau undergoes many modifications, including phosphorylation, arginine monomethylation, lysine acetylation, lysine monomethylation, lysine dimethylation, lysine ubiquitylation and serine.
O-linked N-acetylglucosamine (O-GlcNAc) modification [39]. Under pathological conditions, increasing of tau hyperphosphorylation will render the protein aggregation-proned, reduce its affinity for microtubules, and thereby influence neuronal plasticity. Consequently, strategies to target tau involve blocking of tau aggregation, utilizing tau vaccinations, stabilizing microtubules, manipulating kinases and phosphatases that govern tau modifications. However, most of these efforts have failed in clinical trials. For Tau aggregation blockers, TRx0237 failed to show treatment benefits in phase III trials [40]. As for vaccinations, tau-targeted active vaccines (ACI35 and AADvac-1) and passive vaccines (RG6100 and ABBv-8E12) are currently in phase I and II clinical trials [41, 42]. Intravenous immunoglobulin (IVIG), the only passive vaccine in phase III clinical trials, failed to meet the primary end points in patients with mild-to-moderate AD [42]. Other tau-targeting strategies for AD, including stabilizing microtubules and manipulating kinases and phosphatases, have just been tested in preclinical studies.
In general, tau-targeting therapies remain challenging because of incomplete understanding of AD, lack of robust and sensitive biomarkers for diagnosis and response-monitoring, and the obstruction of blood-brain barrier.

Inflammation hypothesis

Reactive gliosis and neuroinflammation are hallmarks of AD. Microglia-related pathways were considered to be central to AD risk and pathogenesis, as supported by emerging genetic and transcriptomic studies [4347]. Increasing evidence demonstrate that microglia emerges as central players in AD. In very early stage, microglia, TREM2 and complement system are responsible for synaptic pruning [48, 49]. The processes of activity-dependent and long-term synaptic plasticity are the common and fundamental cellular underpinning of learning and memory which may manifest as influence on long term potential [50]. Following that, reactive microglia and astrocytes will surround amyloid plaques and secrete numerous pro-inflammatory cytokines. These events are regarded as an early, prime mover in AD evolution. However, non-steroid anti-inflammatory drugs (NSAIDs) did not show enough benefits in clinic. This is because that the relationship between innate immunity and AD pathogenesis is complex, and the immune response can be either deleterious or beneficial depending on the context [47, 51, 52]. However, the new observations that PD-1 immune checkpoint blockade reduces the pathology of AD and improves memory in mouse models of AD [5355] give us a direction of future researches.
The recent advances in our understanding of the mechanism underlying microglia dysfunction in pruning, regulating plasticity, and neurogenesis are opening up possibilities for new opportunities of AD therapeutic interventions and diagnosis [56, 57]. Targeting these aberrant microglial functions and thereby returning homeostasis may yield novel paradigms for AD therapies. However, given the complexity and diverse functions of microglia in health and disease, there is a crucial need for new biomarkers reflecting the function of specific microglias [52, 58].

Cholinergic and oxidative stress hypothesis

Acetylcholine (ACh) is an important neurotransmitter used by cholinergic neurons, which has been involved in critical physiological processes, such as attention, learning, memory, stress response, wakefulness and sleep, and sensory information [5963]. Cholinergic neurons damage was considered to be a critical pathological change that correlated with cognitive impairment in AD. Thus, cholinergic hypothesis was firstly tested with cholinesterase inhibitors in AD treatment. Tacrine, a cholinesterase inhibitor, was the first anti-AD drug available in clinic [6466] although it was withdrawn from the market in 2012 due to severe side effects. Although inhibiting cholinesterase is a symptomatic relief treatment with marginal benefits, it is currently the most available clinical treatment which gives desperate AD patients a glimmer of hope. For other neurotransmitter dysfunction, such as Dopamine and 5-hydroxytryptamine, there are some studies about them, but not much as acetylcholine in AD.
Oxidative stress is considered to play an important role in the pathogenesis of AD. Especially, the brain utilizes more oxygen than other tissues and undergoes mitochondrial respiration, which increases the potential for ROS exposure. In fact, AD is highly associated with cellular oxidative stress, including augmentation of protein oxidation, protein nitration, glycoloxidation and lipid peroxidation as well as accumulation of Aβ, for Aβ can also induce oxidative stress [6773]. Thus, the treatment with anti-oxidant compounds would provide protection against oxidative stress and Aβ toxicity in theory. However, oxidative stress is only a single feature of AD, so antioxidant strategy was challenged for its potency to stop the progression of AD and thus it is proposed as a portion of combination therapy [74, 75].

Glucose hypometabolism

Glucose hypometabolism is the early pathogenic event in the prodromal phase of AD, and associated with cognitive and functional decline. Early therapeutic intervention before the irreversible degeneration has become a consensus in AD treatment. Thus, alleviation of glucose hypometabolism was emerged as an attractive strategy of AD treatment. However, most of these therapeutic strategies are targeting mitochondria and bioenergetics, which have shown promise at the preclinical stage but without success in clinical trials [76, 77]. Although no strategies are available to alleviate glucose hypometabolism in clinical, glucose metabolism brain imaging such as 18FDG-PET (Positron emision tomography with 2-deoxy-2-fluorine-18-fluoro-D-glucose) has become a valuable indicator for diagnosis of neurodegenerative diseases that cause dementia, including AD [78].
Up to now, there’re no effective treatments for changing the course of AD. Confronting these difficulties, we should get deeper understandings about these hypotheses, and meanwhile we should renovate our knowledge about AD and develop new hypothesis.

New pathway to AD

AD is conventionally regarded as a central nervous system (CNS) disorder. However, increasing experimental, epidemiological and clinical evidences have suggested that manifestations of AD extend beyond the brain. Most notably, research over the past few years reveals that the gut microbiome (GMB) has a profound impact on the formation of the blood-brain barrier, myelination, neurogenesis, and microglia maturation [7984]. In particular, results from germ-free animals and animals exposed to pathogenic microbial infections, antibiotics, probiotics, or fecal microbiota transplantation showed that gut microbiota modulates many aspects of animal behaviors, suggesting a role for the gut microbiota in host cognition or AD-related pathogenesis [8588]. The underlying mechanisms of gut microbiota influencing brain involve the communication through immune system, the endocrine system, the vague nerve, and the bacteria-derived metabolites.

Immune pathway

The intestinal mucosal lymphoid tissue contains 70% ~ 80% of the immune cells in the whole body, and is considered to be the largest and most important human immune organs. It is also the first line of host defense against pathogens. The human gut contains a large, diverse and dynamic enteric microbiota, including more than 100 trillion microorganisms from at least 1000 distinct species. There’s a complex relationship between intestinal mucosal immune system and intestinal microbiota. Thus, gut microbiota induced immunomodulation is emerging as an important pathway that influences AD [89].
Gut microbiota can influence brain through immune system in several ways. Firstly, intestinal microbiome can induce cytokines secretion, which enter the circulatory system, pass through blood brain barrier, and directly affect the brain function. For instance, perivascular macrophages and cerebral small vessel epithelial cells can receive the intestinal microbiome produced IL-1 signal and affect central nervous system. Also, gut microbes can activate Toll-like receptors of the brain immune cells (such as microglia) through microbes associated molecular patterns (MAMP). MAMPs can either directly bind to intestinal epithelial cells or infiltrate to the intestine lamina propria to activate lymphocytes, promoting the release of pro-inflammatory cytokines, which further cause subsequent inflammation in brain. Secondly, gut microbes can produce metabolites such as short-chain-fatty acids (SCFAs), gamma-aminobutyric acid (GABA) and 5-HT precursors, which could also travel to the brain via circulatory systems or signal through intestinal epithelials to produce cytokines or neurotransmitters that activate vagus nerve. Thirdly, gut microbes can activate enteroendocrine cells to produce 5-HT, which affect the brain through neuroimmune pathways.
In addition to changing the functions of the immune system, such as through secretion of inflammatory factors or anti-inflammatory factors, intestinal microbiome can also affect the development and composition of immune system. For example, in germ-free mice, isolated lymphoid follicles in gut associated lymphoid tissue are unable to mature, and lymphocytes that are able to secrete IgA in the intestinal epithelium decreased [8992]. For immune system in brain, the deletion of gut microbiota in germ-free mice have global influence on the cell proportions and maturation of microglia in the brain, and thus affect the properties and phenotype of microglia, as compared to conventionally colonized controls [93]. Similar results were obtained in antibiotic treated mice. Other research also demonstrates that the number of T regulatory cells and T helper lymphocytes (T helper 17, Th17) are significantly reduced in the germ free mouse, indicating the regulatory effects of intestinal microbiome on T cell composition, while microbiome tansplant to germ free mice can modify these variations and restore normal immune function [94, 95]. All these modulations of gut microbiota may have direct and indirect effects on AD development and progression.

Endocrine pathway and the vagus nerve

The gut is also the largest endocrine organ in the body. Gut microbiota can regulate secretion of many hormones from intestinal endocrine cells, such as corticosterone and adrenal hormones, and thus establish the information exchange between the intestines and the brain. For example, the intestinal microbiome can affect the secretion of serotonin and regulate brain emotional activities [96, 97]; intestinal microbial metabolism can also produce a variety of neurotransmitters, such as dopamine, GABA, acetylcholine and melatonin, which are transmitted to central nervous system through the vagus nerve [98]. Besides transporting these signal substances, the vagus nerve itself plays an important role in inflammation and depression [99]. The vagus nerve can influence the gastrointestinal tract, orchestrate the complex interactions between central and peripheral neural control mechanisms [100]. The stimulation of vagus nerve is able to regulate mood, and the immune system, suggesting the therapeutic potential of vagus nerve modulation to attenuate the pathophysiological changes and restore homeostasis [98103].

Bacteria-derived metabolites

Generation of essential nutrients for host physiology, such as vitamins and other cofactors, is an important physiological function of the gut microbiota [104]. The metabolites of microbiome, such as SCFAs including acetate, butyrate, and propionate, are able to modulate peripheral and central pathologic processes [105]. For example, butyrate is effective in reducing inflammation and pain. Once in the brain, acetate is able to alter the level of the neurotransmitters glutamate, glutamine, and GABA, as well as increases anorectic neuropeptide expression [106]. In addition, the gut microbiota can secrete large amounts of amyloids and lipopolysaccharides, which might contribute to the modulation of signaling pathways, the production of proinflammatory cytokines associated with AD pathogenesis and Aβ deposition [107109].
In fact, microbiota-gut-brain axis has been established and a disturbed gut microbiota has been incriminated in many neurodegenerative diseases in animal and translational models. In theory, a role for the microbiota-gut-brain axis is highly plausible. However, the theoretical basis for the use of microbiota-directed therapies in neurodegenerative disorders still needs supports from high-quality clinical trials [110]. To date, only a few studies directly focused on the gut microbiota and AD [111, 112], and studies on AD patients is particullarly deficient. A recent research from human showed an increase in the abundance of a pro-inflammatory GMB taxon and a reduction in the abundance of an anti-inflammatory taxon are possibly associated with a peripheral inflammatory state in patients with cognitive impairment and brain amyloidosis. It is important for the research of gut microbiota and AD. However, further investigations are still necessary to explore the possible causal relation between GMB-related inflammation and amyloidosis [111]. The comprehensive understanding of these underlying mechanisms may provide new insights into these novel therapeutic strategies for AD. In particular, based on the gut microbiota hypothesis, Chinese traditional medicine and probiotic bacteria may play a more important role in therapy [113].

Conclusions

Nowadays, new technologies are making it possible to get to know enough pathologic details of disease. More importantly, scientists are beginning to treat AD as a systemic disease and they are paying more attention to the correlation between brain and other organs [47, 89, 114]. Perhaps, for complicated disease such as AD, researches and therapies should be based on the principle that combined reductionism with holism, and great efforts should be made to search the fundamental laws of AD by means of multi-scale modeling and efficient numeric assessment. Maybe, just like Chinese traditional medicine [115], combination treatments or systematic therapy will be a final way out.

Acknowledgements

Not applicable.

Funding

This review was supported by “Personalized Medicines-Molecular Signature based Drug Discovery and Development”, Strategic Priority Research program of Sciences, Grants No. XDA12040101.

Availability of data and materials

Not applicable.
Not relevant.
Not applicable.

Competing interests

The authors declare they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Ali G-C, Guerchet M, Wu Y-T, Prince M, Prina M. Chapter 2: The global prevalence of dementia. In: Prince M, Guerchet M, Ali G-C, Wu Y-T, Prina M, editors. The Global Impact of Dementia. An analysis of prevalence, incidence, cost and trends. London: Alzheimer’s Disease International (ADI); 2015. p. 10–29. Ali G-C, Guerchet M, Wu Y-T, Prince M, Prina M. Chapter 2: The global prevalence of dementia. In: Prince M, Guerchet M, Ali G-C, Wu Y-T, Prina M, editors. The Global Impact of Dementia. An analysis of prevalence, incidence, cost and trends. London: Alzheimer’s Disease International (ADI); 2015. p. 10–29.
3.
Zurück zum Zitat Vassar R, Citron M. Abeta-generating enzymes: recent advances in beta- and gamma-secretase research. Neuron. 2000;27(3):419–22.PubMedCrossRef Vassar R, Citron M. Abeta-generating enzymes: recent advances in beta- and gamma-secretase research. Neuron. 2000;27(3):419–22.PubMedCrossRef
4.
Zurück zum Zitat van Es JH, et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 2005;435(7044):959–63.PubMedCrossRef van Es JH, et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 2005;435(7044):959–63.PubMedCrossRef
5.
Zurück zum Zitat Olry A, et al. Generation and characterization of mutant cell lines defective in gamma-secretase processing of notch and amyloid precursor protein. J Biol Chem. 2005;280(31):28564–71.PubMedCrossRef Olry A, et al. Generation and characterization of mutant cell lines defective in gamma-secretase processing of notch and amyloid precursor protein. J Biol Chem. 2005;280(31):28564–71.PubMedCrossRef
6.
Zurück zum Zitat Tarassishin L, et al. Processing of notch and amyloid precursor protein by gamma-secretase is spatially distinct. Proc Natl Acad Sci U S A. 2004;101(49):17050–5.PubMedPubMedCentralCrossRef Tarassishin L, et al. Processing of notch and amyloid precursor protein by gamma-secretase is spatially distinct. Proc Natl Acad Sci U S A. 2004;101(49):17050–5.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Sastre M, et al. Presenilin-dependent gamma-secretase processing of beta-amyloid precursor protein at a site corresponding to the S3 cleavage of notch. EMBO Rep. 2001;2(9):835–41.PubMedPubMedCentralCrossRef Sastre M, et al. Presenilin-dependent gamma-secretase processing of beta-amyloid precursor protein at a site corresponding to the S3 cleavage of notch. EMBO Rep. 2001;2(9):835–41.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Klaver DW, et al. Is BACE1 a suitable therapeutic target for the treatment of Alzheimer’s disease? Current strategies and future directions. Biol Chem. 2010;391(8):849–59.PubMedCrossRef Klaver DW, et al. Is BACE1 a suitable therapeutic target for the treatment of Alzheimer’s disease? Current strategies and future directions. Biol Chem. 2010;391(8):849–59.PubMedCrossRef
9.
Zurück zum Zitat Abramov E, et al. Amyloid-beta as a positive endogenous regulator of release probability at hippocampal synapses. Nat Neurosci. 2009;12(12):1567–76.PubMedCrossRef Abramov E, et al. Amyloid-beta as a positive endogenous regulator of release probability at hippocampal synapses. Nat Neurosci. 2009;12(12):1567–76.PubMedCrossRef
10.
Zurück zum Zitat Gilman S, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in AN interrupted trial. Neurology. 2005;64(9):1553–62.PubMedCrossRef Gilman S, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in AN interrupted trial. Neurology. 2005;64(9):1553–62.PubMedCrossRef
11.
Zurück zum Zitat Bayer AJ, et al. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology. 2005;64(1):94–101.PubMedCrossRef Bayer AJ, et al. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology. 2005;64(1):94–101.PubMedCrossRef
12.
Zurück zum Zitat Holmes C, et al. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372(9634):216–23.PubMedCrossRef Holmes C, et al. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372(9634):216–23.PubMedCrossRef
13.
Zurück zum Zitat Laskowitz DT, Kolls BJ. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2010;74(24):2026. author reply 2026-7PubMedCrossRef Laskowitz DT, Kolls BJ. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2010;74(24):2026. author reply 2026-7PubMedCrossRef
14.
Zurück zum Zitat Salloway S, et al. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2009;73(24):2061–70.PubMedPubMedCentralCrossRef Salloway S, et al. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2009;73(24):2061–70.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Bouter Y, et al. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against Ntruncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol. 2015;130(5):713–29.PubMedCrossRef Bouter Y, et al. Abeta targets of the biosimilar antibodies of Bapineuzumab, Crenezumab, Solanezumab in comparison to an antibody against Ntruncated Abeta in sporadic Alzheimer disease cases and mouse models. Acta Neuropathol. 2015;130(5):713–29.PubMedCrossRef
17.
Zurück zum Zitat The Lancet N. Solanezumab: too late in mild Alzheimer’ss disease? Lancet Neurol. 2017;16(2):97.CrossRef The Lancet N. Solanezumab: too late in mild Alzheimer’ss disease? Lancet Neurol. 2017;16(2):97.CrossRef
18.
Zurück zum Zitat Gandy S, Sano M. Alzheimer disease: Solanezumab-prospects for meaningful interventions in AD? Nat Rev Neurol. 2015;11(12):669–70.PubMedCrossRef Gandy S, Sano M. Alzheimer disease: Solanezumab-prospects for meaningful interventions in AD? Nat Rev Neurol. 2015;11(12):669–70.PubMedCrossRef
19.
Zurück zum Zitat Landen JW, et al. Safety and pharmacology of a single intravenous dose of ponezumab in subjects with mild-to-moderate Alzheimer disease: a phase I, randomized, placebo-controlled, double-blind, dose-escalation study. Clin Neuropharmacol. 2013;36(1):14–23.PubMedCrossRef Landen JW, et al. Safety and pharmacology of a single intravenous dose of ponezumab in subjects with mild-to-moderate Alzheimer disease: a phase I, randomized, placebo-controlled, double-blind, dose-escalation study. Clin Neuropharmacol. 2013;36(1):14–23.PubMedCrossRef
20.
Zurück zum Zitat Burstein AH, et al. Safety and pharmacology of ponezumab (PF-04360365) after a single 10-minute intravenous infusion in subjects with mild to moderate Alzheimer disease. Clin Neuropharmacol. 2013;36(1):8–13.PubMedCrossRef Burstein AH, et al. Safety and pharmacology of ponezumab (PF-04360365) after a single 10-minute intravenous infusion in subjects with mild to moderate Alzheimer disease. Clin Neuropharmacol. 2013;36(1):8–13.PubMedCrossRef
21.
Zurück zum Zitat La Porte SL, et al. Structural basis of C-terminal beta-amyloid peptide binding by the antibody ponezumab for the treatment of Alzheimer's disease. J Mol Biol. 2012;421(4–5):525–36.PubMedCrossRef La Porte SL, et al. Structural basis of C-terminal beta-amyloid peptide binding by the antibody ponezumab for the treatment of Alzheimer's disease. J Mol Biol. 2012;421(4–5):525–36.PubMedCrossRef
22.
Zurück zum Zitat Carlson C, et al. Amyloid-related imaging abnormalities from trials of solanezumab for Alzheimer's disease. Alzheimers Dement (Amst). 2016;2:75–85. Carlson C, et al. Amyloid-related imaging abnormalities from trials of solanezumab for Alzheimer's disease. Alzheimers Dement (Amst). 2016;2:75–85.
23.
Zurück zum Zitat Salloway S, et al. A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease. Neurology. 2011;77(13):1253–62.PubMedPubMedCentralCrossRef Salloway S, et al. A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease. Neurology. 2011;77(13):1253–62.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Aisen PS, et al. Tramiprosate in mild-to-moderate Alzheimer's disease - a randomized, double-blind, placebo-controlled, multi-centre study (the Alphase study). Arch Med Sci. 2011;7(1):102–11.PubMedPubMedCentralCrossRef Aisen PS, et al. Tramiprosate in mild-to-moderate Alzheimer's disease - a randomized, double-blind, placebo-controlled, multi-centre study (the Alphase study). Arch Med Sci. 2011;7(1):102–11.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Greenberg SM, et al. A phase 2 study of tramiprosate for cerebral amyloid angiopathy. Alzheimer Dis Assoc Disord. 2006;20(4):269–74.PubMedCrossRef Greenberg SM, et al. A phase 2 study of tramiprosate for cerebral amyloid angiopathy. Alzheimer Dis Assoc Disord. 2006;20(4):269–74.PubMedCrossRef
26.
Zurück zum Zitat Gervais F, et al. Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol Aging. 2007;28(4):537–47.PubMedCrossRef Gervais F, et al. Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol Aging. 2007;28(4):537–47.PubMedCrossRef
27.
Zurück zum Zitat Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer's disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011;10(9):698–712.PubMedCrossRef Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer's disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011;10(9):698–712.PubMedCrossRef
28.
Zurück zum Zitat Nie Q, Du XG, Geng MY. Small molecule inhibitors of amyloid beta peptide aggregation as a potential therapeutic strategy for Alzheimer's disease. Acta Pharmacol Sin. 2011;32(5):545–51.PubMedPubMedCentralCrossRef Nie Q, Du XG, Geng MY. Small molecule inhibitors of amyloid beta peptide aggregation as a potential therapeutic strategy for Alzheimer's disease. Acta Pharmacol Sin. 2011;32(5):545–51.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Sevigny J, et al. Addendum: the antibody aducanumab reduces Abeta plaques in Alzheimer's disease. Nature. 2017;546(7659):564.PubMedCrossRef Sevigny J, et al. Addendum: the antibody aducanumab reduces Abeta plaques in Alzheimer's disease. Nature. 2017;546(7659):564.PubMedCrossRef
30.
Zurück zum Zitat Sevigny J, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer's disease. Nature. 2016;537(7618):50–6.PubMedCrossRef Sevigny J, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer's disease. Nature. 2016;537(7618):50–6.PubMedCrossRef
31.
Zurück zum Zitat Patel KR. Biogen's aducanumab raises hope that Alzheimer's can be treated at its source. Manag Care. 2015;24(6):19. Patel KR. Biogen's aducanumab raises hope that Alzheimer's can be treated at its source. Manag Care. 2015;24(6):19.
32.
Zurück zum Zitat Karran E. Recent trial shows that solanezumab has disease modifying effects. BMJ. 2015;351:h4528.PubMedCrossRef Karran E. Recent trial shows that solanezumab has disease modifying effects. BMJ. 2015;351:h4528.PubMedCrossRef
33.
Zurück zum Zitat Siemers ER, et al. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer's disease patients. Alzheimers Dement. 2016;12(2):110–20.PubMedCrossRef Siemers ER, et al. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer's disease patients. Alzheimers Dement. 2016;12(2):110–20.PubMedCrossRef
34.
Zurück zum Zitat Barrera-Ocampo A, Lopera F. Amyloid-beta immunotherapy: the hope for Alzheimer disease? Colomb Med (Cali). 2016;47(4):203–12. Barrera-Ocampo A, Lopera F. Amyloid-beta immunotherapy: the hope for Alzheimer disease? Colomb Med (Cali). 2016;47(4):203–12.
35.
Zurück zum Zitat Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5.PubMedCrossRef Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5.PubMedCrossRef
37.
Zurück zum Zitat Wang J, et al. A systemic view of Alzheimer disease - insights from amyloid-beta metabolism beyond the brain. Nat Rev Neurol. 2017;13(10):612–23.PubMedCrossRef Wang J, et al. A systemic view of Alzheimer disease - insights from amyloid-beta metabolism beyond the brain. Nat Rev Neurol. 2017;13(10):612–23.PubMedCrossRef
39.
Zurück zum Zitat Morris M, et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci. 2015;18(8):1183–9.PubMedCrossRef Morris M, et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci. 2015;18(8):1183–9.PubMedCrossRef
40.
Zurück zum Zitat Gauthier S, et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer's disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet. 2016;388(10062):2873–84.PubMedPubMedCentralCrossRef Gauthier S, et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer's disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet. 2016;388(10062):2873–84.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Novak P, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer's disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol. 2017;16(2):123–34.PubMedCrossRef Novak P, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer's disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol. 2017;16(2):123–34.PubMedCrossRef
42.
Zurück zum Zitat Li C, Gotz J. Tau-based therapies in neurodegeneration: opportunities and challenges. Nat Rev Drug Discov. 2017; Li C, Gotz J. Tau-based therapies in neurodegeneration: opportunities and challenges. Nat Rev Drug Discov. 2017;
43.
44.
Zurück zum Zitat Guerreiro R, et al. TREM2 variants in Alzheimer's disease. N Engl J Med. 2013;368(2):117–27.PubMedCrossRef Guerreiro R, et al. TREM2 variants in Alzheimer's disease. N Engl J Med. 2013;368(2):117–27.PubMedCrossRef
45.
Zurück zum Zitat Song W, et al. Alzheimer's disease-associated TREM2 variants exhibit either decreased or increased ligand-dependent activation. Alzheimers Dement. 2017;13(4):381–7.PubMedCrossRef Song W, et al. Alzheimer's disease-associated TREM2 variants exhibit either decreased or increased ligand-dependent activation. Alzheimers Dement. 2017;13(4):381–7.PubMedCrossRef
46.
Zurück zum Zitat Colonna M, Wang Y. TREM2 variants: new keys to decipher Alzheimer disease pathogenesis. Nat Rev Neurosci. 2016;17(4):201–7.PubMedCrossRef Colonna M, Wang Y. TREM2 variants: new keys to decipher Alzheimer disease pathogenesis. Nat Rev Neurosci. 2016;17(4):201–7.PubMedCrossRef
47.
Zurück zum Zitat Bolos M, Perea JR, Avila J. Alzheimer's disease as an inflammatory disease. Biomol Concepts. 2017;8(1):37–43.PubMedCrossRef Bolos M, Perea JR, Avila J. Alzheimer's disease as an inflammatory disease. Biomol Concepts. 2017;8(1):37–43.PubMedCrossRef
48.
Zurück zum Zitat Hong S, Dissing-Olesen L, Stevens B. New insights on the role of microglia in synaptic pruning in health and disease. Curr Opin Neurobiol. 2016;36:128–34.PubMedCrossRef Hong S, Dissing-Olesen L, Stevens B. New insights on the role of microglia in synaptic pruning in health and disease. Curr Opin Neurobiol. 2016;36:128–34.PubMedCrossRef
49.
Zurück zum Zitat Paolicelli RC, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333(6048):1456–8.PubMedCrossRef Paolicelli RC, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333(6048):1456–8.PubMedCrossRef
50.
Zurück zum Zitat Bliss TV, Collingridge GL, Morris RG. Synaptic plasticity in health and disease: introduction and overview. Philos Trans R Soc Lond Ser B Biol Sci. 2014;369(1633):20130129.CrossRef Bliss TV, Collingridge GL, Morris RG. Synaptic plasticity in health and disease: introduction and overview. Philos Trans R Soc Lond Ser B Biol Sci. 2014;369(1633):20130129.CrossRef
51.
Zurück zum Zitat Chen J, et al. Inhibition of AGEs/RAGE/rho/ROCK pathway suppresses non-specific neuroinflammation by regulating BV2 microglial M1/M2 polarization through the NF-kappaB pathway. J Neuroimmunol. 2017;305:108–14.PubMedCrossRef Chen J, et al. Inhibition of AGEs/RAGE/rho/ROCK pathway suppresses non-specific neuroinflammation by regulating BV2 microglial M1/M2 polarization through the NF-kappaB pathway. J Neuroimmunol. 2017;305:108–14.PubMedCrossRef
52.
Zurück zum Zitat Hirbec H E, Noristani HN, Perrin FE. Microglia Responses in Acute and Chronic Neurological Diseases: What Microglia-Specific Transcriptomic Studies Taught (and did Not Teach) Us. Front Aging Neurosci. 2017;9:227.CrossRef Hirbec H E, Noristani HN, Perrin FE. Microglia Responses in Acute and Chronic Neurological Diseases: What Microglia-Specific Transcriptomic Studies Taught (and did Not Teach) Us. Front Aging Neurosci. 2017;9:227.CrossRef
53.
Zurück zum Zitat Baruch K, et al. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer's disease. Nat Med. 2016;22(2):135–7.PubMedCrossRef Baruch K, et al. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer's disease. Nat Med. 2016;22(2):135–7.PubMedCrossRef
54.
Zurück zum Zitat Saresella M, et al. A potential role for the PD1/PD-L1 pathway in the neuroinflammation of Alzheimer's disease. Neurobiol Aging. 2012;33(3):624 e11–22.CrossRef Saresella M, et al. A potential role for the PD1/PD-L1 pathway in the neuroinflammation of Alzheimer's disease. Neurobiol Aging. 2012;33(3):624 e11–22.CrossRef
55.
Zurück zum Zitat Saresella M, et al. PD1 negative and PD1 positive CD4+ T regulatory cells in mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2010;21(3):927–38.PubMedCrossRef Saresella M, et al. PD1 negative and PD1 positive CD4+ T regulatory cells in mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2010;21(3):927–38.PubMedCrossRef
56.
Zurück zum Zitat Jevtic S, et al. The role of the immune system in Alzheimer disease: etiology and treatment. Ageing Res Rev. 2017;40:84–94.PubMedCrossRef Jevtic S, et al. The role of the immune system in Alzheimer disease: etiology and treatment. Ageing Res Rev. 2017;40:84–94.PubMedCrossRef
57.
Zurück zum Zitat McGeer PL, McGeer EG. Targeting microglia for the treatment of Alzheimer's disease. Expert Opin Ther Targets. 2015;19(4):497–506.PubMedCrossRef McGeer PL, McGeer EG. Targeting microglia for the treatment of Alzheimer's disease. Expert Opin Ther Targets. 2015;19(4):497–506.PubMedCrossRef
58.
Zurück zum Zitat Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017;23(9):1018–27.PubMedCrossRef Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017;23(9):1018–27.PubMedCrossRef
59.
Zurück zum Zitat Hasselmo ME, Anderson BP, Bower JM. Cholinergic modulation of cortical associative memory function. J Neurophysiol. 1992;67(5):1230–46.PubMedCrossRef Hasselmo ME, Anderson BP, Bower JM. Cholinergic modulation of cortical associative memory function. J Neurophysiol. 1992;67(5):1230–46.PubMedCrossRef
60.
Zurück zum Zitat Fine A, et al. Learning impairments following injection of a selective cholinergic immunotoxin, ME20.4 IgG-saporin, into the basal nucleus of Meynert in monkeys. Neuroscience. 1997;81(2):331–43.PubMedCrossRef Fine A, et al. Learning impairments following injection of a selective cholinergic immunotoxin, ME20.4 IgG-saporin, into the basal nucleus of Meynert in monkeys. Neuroscience. 1997;81(2):331–43.PubMedCrossRef
61.
Zurück zum Zitat Sarter M, Bruno JP. Cognitive functions of cortical acetylcholine: toward a unifying hypothesis. Brain Res Brain Res Rev. 1997;23(1–2):28–46.PubMedCrossRef Sarter M, Bruno JP. Cognitive functions of cortical acetylcholine: toward a unifying hypothesis. Brain Res Brain Res Rev. 1997;23(1–2):28–46.PubMedCrossRef
62.
Zurück zum Zitat Miranda MI, Bermudez-Rattoni F. Reversible inactivation of the nucleus basalis magnocellularis induces disruption of cortical acetylcholine release and acquisition, but not retrieval, of aversive memories. Proc Natl Acad Sci U S A. 1999;96(11):6478–82.PubMedPubMedCentralCrossRef Miranda MI, Bermudez-Rattoni F. Reversible inactivation of the nucleus basalis magnocellularis induces disruption of cortical acetylcholine release and acquisition, but not retrieval, of aversive memories. Proc Natl Acad Sci U S A. 1999;96(11):6478–82.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Haam J, Yakel JL. Cholinergic modulation of the hippocampal region and memory function. J Neurochem. 2017;142(Suppl 2):111–21.PubMedCrossRef Haam J, Yakel JL. Cholinergic modulation of the hippocampal region and memory function. J Neurochem. 2017;142(Suppl 2):111–21.PubMedCrossRef
64.
Zurück zum Zitat Brinkman SD, Gershon S. Measurement of cholinergic drug effects on memory in Alzheimer's disease. Neurobiol Aging. 1983;4(2):139–45.PubMedCrossRef Brinkman SD, Gershon S. Measurement of cholinergic drug effects on memory in Alzheimer's disease. Neurobiol Aging. 1983;4(2):139–45.PubMedCrossRef
65.
Zurück zum Zitat Summers WK, et al. Oral tetrahydroaminoacridine in long-term treatment of senile dementia, Alzheimer type. N Engl J Med. 1986;315(20):1241–5.PubMedCrossRef Summers WK, et al. Oral tetrahydroaminoacridine in long-term treatment of senile dementia, Alzheimer type. N Engl J Med. 1986;315(20):1241–5.PubMedCrossRef
66.
Zurück zum Zitat Summers WK, et al. Use of THA in treatment of Alzheimer-like dementia: pilot study in twelve patients. Biol Psychiatry. 1981;16(2):145–53.PubMed Summers WK, et al. Use of THA in treatment of Alzheimer-like dementia: pilot study in twelve patients. Biol Psychiatry. 1981;16(2):145–53.PubMed
67.
Zurück zum Zitat Cheignon C, et al. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol. 2018;14:450–64.PubMedCrossRef Cheignon C, et al. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol. 2018;14:450–64.PubMedCrossRef
68.
Zurück zum Zitat Sultana R, Butterfield DA. Redox proteomics studies of in vivo amyloid beta-peptide animal models of Alzheimer's disease: insight into the role of oxidative stress. Proteomics Clin Appl. 2008;2(5):685–96.PubMedCrossRef Sultana R, Butterfield DA. Redox proteomics studies of in vivo amyloid beta-peptide animal models of Alzheimer's disease: insight into the role of oxidative stress. Proteomics Clin Appl. 2008;2(5):685–96.PubMedCrossRef
69.
Zurück zum Zitat Butterfield DA, et al. Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer's disease and mild cognitive impairment. Free Radic Biol Med. 2007;43(5):658–77.PubMedPubMedCentralCrossRef Butterfield DA, et al. Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer's disease and mild cognitive impairment. Free Radic Biol Med. 2007;43(5):658–77.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Mohmmad Abdul H, et al. Mutations in amyloid precursor protein and presenilin-1 genes increase the basal oxidative stress in murine neuronal cells and lead to increased sensitivity to oxidative stress mediated by amyloid beta-peptide (1-42), HO and kainic acid: implications for Alzheimer's disease. J Neurochem. 2006;96(5):1322–35.PubMedCrossRef Mohmmad Abdul H, et al. Mutations in amyloid precursor protein and presenilin-1 genes increase the basal oxidative stress in murine neuronal cells and lead to increased sensitivity to oxidative stress mediated by amyloid beta-peptide (1-42), HO and kainic acid: implications for Alzheimer's disease. J Neurochem. 2006;96(5):1322–35.PubMedCrossRef
71.
Zurück zum Zitat Gibson GL, Allsop D, Austen BM. Induction of cellular oxidative stress by the beta-amyloid peptide involved in Alzheimer's disease. Protein Pept Lett. 2004;11(3):257–70.PubMedCrossRef Gibson GL, Allsop D, Austen BM. Induction of cellular oxidative stress by the beta-amyloid peptide involved in Alzheimer's disease. Protein Pept Lett. 2004;11(3):257–70.PubMedCrossRef
72.
Zurück zum Zitat Butterfield DA, et al. Amyloid beta-peptide and amyloid pathology are central to the oxidative stress and inflammatory cascades under which Alzheimer's disease brain exists. J Alzheimers Dis. 2002;4(3):193–201.PubMedCrossRef Butterfield DA, et al. Amyloid beta-peptide and amyloid pathology are central to the oxidative stress and inflammatory cascades under which Alzheimer's disease brain exists. J Alzheimers Dis. 2002;4(3):193–201.PubMedCrossRef
73.
Zurück zum Zitat Butterfield DA, Lauderback CM. Lipid peroxidation and protein oxidation in Alzheimer's disease brain: potential causes and consequences involving amyloid beta-peptide-associated free radical oxidative stress. Free Radic Biol Med. 2002;32(11):1050–60.PubMedCrossRef Butterfield DA, Lauderback CM. Lipid peroxidation and protein oxidation in Alzheimer's disease brain: potential causes and consequences involving amyloid beta-peptide-associated free radical oxidative stress. Free Radic Biol Med. 2002;32(11):1050–60.PubMedCrossRef
74.
Zurück zum Zitat Persson T, Popescu BO, Cedazo-Minguez A. Oxidative stress in Alzheimer's disease: why did antioxidant therapy fail? Oxidative Med Cell Longev. 2014;2014:427318.CrossRef Persson T, Popescu BO, Cedazo-Minguez A. Oxidative stress in Alzheimer's disease: why did antioxidant therapy fail? Oxidative Med Cell Longev. 2014;2014:427318.CrossRef
75.
Zurück zum Zitat Teixeira J, et al. Alzheimer's disease and antioxidant therapy: how long how far? Curr Med Chem. 2013;20(24):2939–52.PubMedCrossRef Teixeira J, et al. Alzheimer's disease and antioxidant therapy: how long how far? Curr Med Chem. 2013;20(24):2939–52.PubMedCrossRef
76.
Zurück zum Zitat Caldwell CC, Yao J, Brinton RD. Targeting the prodromal stage of Alzheimer's disease: bioenergetic and mitochondrial opportunities. Neurotherapeutics. 2015;12(1):66–80.PubMedCrossRef Caldwell CC, Yao J, Brinton RD. Targeting the prodromal stage of Alzheimer's disease: bioenergetic and mitochondrial opportunities. Neurotherapeutics. 2015;12(1):66–80.PubMedCrossRef
77.
Zurück zum Zitat Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res. 2017;95(4):943–72.PubMedCrossRef Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res. 2017;95(4):943–72.PubMedCrossRef
78.
Zurück zum Zitat Nasrallah I, Dubroff J. An overview of PET neuroimaging. Semin Nucl Med. 2013;43(6):449–61.PubMedCrossRef Nasrallah I, Dubroff J. An overview of PET neuroimaging. Semin Nucl Med. 2013;43(6):449–61.PubMedCrossRef
79.
80.
Zurück zum Zitat Stephan AH, Barres BA, Stevens B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89.PubMedCrossRef Stephan AH, Barres BA, Stevens B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89.PubMedCrossRef
81.
Zurück zum Zitat Lui H, et al. Progranulin deficiency promotes circuit-specific synaptic pruning by microglia via complement activation. Cell. 2016;165(4):921–35.PubMedPubMedCentralCrossRef Lui H, et al. Progranulin deficiency promotes circuit-specific synaptic pruning by microglia via complement activation. Cell. 2016;165(4):921–35.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Elmer BM, McAllister AK. Major histocompatibility complex class I proteins in brain development and plasticity. Trends Neurosci. 2012;35(11):660–70.PubMedPubMedCentralCrossRef Elmer BM, McAllister AK. Major histocompatibility complex class I proteins in brain development and plasticity. Trends Neurosci. 2012;35(11):660–70.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Erickson MA, Dohi K, Banks WA. Neuroinflammation: a common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation. 2012;19(2):121–30.PubMedPubMedCentralCrossRef Erickson MA, Dohi K, Banks WA. Neuroinflammation: a common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation. 2012;19(2):121–30.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Banks WA. The blood-brain barrier in neuroimmunology: Tales of separation and assimilation. Brain Behav Immun. 2015;44:1–8.PubMedCrossRef Banks WA. The blood-brain barrier in neuroimmunology: Tales of separation and assimilation. Brain Behav Immun. 2015;44:1–8.PubMedCrossRef
85.
Zurück zum Zitat Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol. 2012;10(11):735–42.PubMedCrossRef Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol. 2012;10(11):735–42.PubMedCrossRef
86.
Zurück zum Zitat Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012;13(10):701–12.PubMedCrossRef Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012;13(10):701–12.PubMedCrossRef
87.
89.
Zurück zum Zitat Belkaid Y, Harrison OJ. Homeostatic immunity and the microbiota. Immunity. 2017;46(4):562–76.PubMedCrossRef Belkaid Y, Harrison OJ. Homeostatic immunity and the microbiota. Immunity. 2017;46(4):562–76.PubMedCrossRef
91.
Zurück zum Zitat Tomkovich S, Jobin C. Microbiota and host immune responses: a love-hate relationship. Immunology. 2016;147(1):1–10.PubMedCrossRef Tomkovich S, Jobin C. Microbiota and host immune responses: a love-hate relationship. Immunology. 2016;147(1):1–10.PubMedCrossRef
92.
Zurück zum Zitat Agace WW, McCoy KD. Regionalized development and maintenance of the intestinal adaptive immune landscape. Immunity. 2017;46(4):532–48.PubMedCrossRef Agace WW, McCoy KD. Regionalized development and maintenance of the intestinal adaptive immune landscape. Immunity. 2017;46(4):532–48.PubMedCrossRef
93.
95.
Zurück zum Zitat Sudo N, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558(Pt 1):263–75.PubMedPubMedCentralCrossRef Sudo N, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558(Pt 1):263–75.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat O'Mahony SM, et al. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:32–48.PubMedCrossRef O'Mahony SM, et al. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:32–48.PubMedCrossRef
97.
Zurück zum Zitat Praveen V, Praveen S. Microbiome-gut-brain Axis: a pathway for improving brainstem serotonin homeostasis and successful autoresuscitation in SIDS-A novel hypothesis. Front Pediatr. 2016;4:136.PubMed Praveen V, Praveen S. Microbiome-gut-brain Axis: a pathway for improving brainstem serotonin homeostasis and successful autoresuscitation in SIDS-A novel hypothesis. Front Pediatr. 2016;4:136.PubMed
98.
Zurück zum Zitat Jiang XG, et al. Research progress in anti-inflammation of vagus nerve and neurotransmitter ach. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2003;15(1):59–61.PubMed Jiang XG, et al. Research progress in anti-inflammation of vagus nerve and neurotransmitter ach. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2003;15(1):59–61.PubMed
99.
Zurück zum Zitat Das UN. Vagus nerve stimulation, depression, and inflammation. Neuropsychopharmacology. 2007;32(9):2053–4.PubMedCrossRef Das UN. Vagus nerve stimulation, depression, and inflammation. Neuropsychopharmacology. 2007;32(9):2053–4.PubMedCrossRef
100.
Zurück zum Zitat Browning KN, Verheijden S, Boeckxstaens GE. The Vagus nerve in appetite regulation, mood, and intestinal inflammation. Gastroenterology. 2017;152(4):730–44.PubMedCrossRef Browning KN, Verheijden S, Boeckxstaens GE. The Vagus nerve in appetite regulation, mood, and intestinal inflammation. Gastroenterology. 2017;152(4):730–44.PubMedCrossRef
101.
Zurück zum Zitat de Haan JJ, et al. Exploring the link between inflammation and the vagus nerve. J Intern Med. 2010;267(1):130–1.PubMedCrossRef de Haan JJ, et al. Exploring the link between inflammation and the vagus nerve. J Intern Med. 2010;267(1):130–1.PubMedCrossRef
102.
Zurück zum Zitat Hoeger S, et al. Modulation of brain dead induced inflammation by vagus nerve stimulation. Am J Transplant. 2010;10(3):477–89.PubMedCrossRef Hoeger S, et al. Modulation of brain dead induced inflammation by vagus nerve stimulation. Am J Transplant. 2010;10(3):477–89.PubMedCrossRef
104.
Zurück zum Zitat Gordon JI, et al. The human gut microbiota and undernutrition. Sci Transl Med. 2012;4(137):137ps12.PubMedCrossRef Gordon JI, et al. The human gut microbiota and undernutrition. Sci Transl Med. 2012;4(137):137ps12.PubMedCrossRef
105.
Zurück zum Zitat Mitchell RW, et al. Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells. J Neurochem. 2011;117(4):735–46.PubMed Mitchell RW, et al. Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells. J Neurochem. 2011;117(4):735–46.PubMed
107.
Zurück zum Zitat Zhao Y, Dua P, Lukiw WJ. Microbial sources of amyloid and relevance to Amyloidogenesis and Alzheimer's disease (AD). J Alzheimers Dis Parkinsonism. 2015;5(1):177.PubMedPubMedCentral Zhao Y, Dua P, Lukiw WJ. Microbial sources of amyloid and relevance to Amyloidogenesis and Alzheimer's disease (AD). J Alzheimers Dis Parkinsonism. 2015;5(1):177.PubMedPubMedCentral
110.
Zurück zum Zitat Quigley EMM. Microbiota-brain-gut Axis and neurodegenerative diseases. Curr Neurol Neurosci Rep. 2017;17(12):94.PubMedCrossRef Quigley EMM. Microbiota-brain-gut Axis and neurodegenerative diseases. Curr Neurol Neurosci Rep. 2017;17(12):94.PubMedCrossRef
111.
Zurück zum Zitat Cattaneo A, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:60–8.PubMedCrossRef Cattaneo A, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017;49:60–8.PubMedCrossRef
112.
Zurück zum Zitat Park AM, et al. Helicobacter pylori and gut microbiota in multiple sclerosis versus Alzheimer's disease: 10 pitfalls of microbiome studies. Clin Exp Neuroimmunol. 2017;8(3):215–32.PubMedCrossRef Park AM, et al. Helicobacter pylori and gut microbiota in multiple sclerosis versus Alzheimer's disease: 10 pitfalls of microbiome studies. Clin Exp Neuroimmunol. 2017;8(3):215–32.PubMedCrossRef
113.
Zurück zum Zitat Chen F, et al. Could the gut microbiota reconcile the oral bioavailability conundrum of traditional herbs? J Ethnopharmacol. 2016;179:253–64.PubMedCrossRef Chen F, et al. Could the gut microbiota reconcile the oral bioavailability conundrum of traditional herbs? J Ethnopharmacol. 2016;179:253–64.PubMedCrossRef
114.
Zurück zum Zitat Zheng X, et al. Thinking outside the brain for cognitive improvement: Is peripheral immunomodulation on the way? Neuropharmacology. 2015;96(Pt A):94–104.PubMedCrossRef Zheng X, et al. Thinking outside the brain for cognitive improvement: Is peripheral immunomodulation on the way? Neuropharmacology. 2015;96(Pt A):94–104.PubMedCrossRef
115.
Zurück zum Zitat Yang WT, et al. Chinese herbal medicine for Alzheimer's disease: clinical evidence and possible mechanism of neurogenesis. Biochem Pharmacol. 2017;141:143–55.PubMedCrossRef Yang WT, et al. Chinese herbal medicine for Alzheimer's disease: clinical evidence and possible mechanism of neurogenesis. Biochem Pharmacol. 2017;141:143–55.PubMedCrossRef
Metadaten
Titel
Alzheimer’s disease hypothesis and related therapies
verfasst von
Xiaoguang Du
Xinyi Wang
Meiyu Geng
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Translational Neurodegeneration / Ausgabe 1/2018
Elektronische ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-018-0107-y

Weitere Artikel der Ausgabe 1/2018

Translational Neurodegeneration 1/2018 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.