Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar
Reach Us +44 1223 790975

GET THE APP

Journal of Alzheimers Disease & Parkinsonism - Alzheimer’s Disease: Molecular Hallmarks and Yeast Models
ISSN: 2161-0460

Journal of Alzheimers Disease & Parkinsonism
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)

Alzheimer’s Disease: Molecular Hallmarks and Yeast Models

Tatiana Goleva, Anton Rogov and Renata Zvyagilskaya*
Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
*Corresponding Author: Renata Zvyagilskaya, Laboratory of Bioenergetics, Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, 33, Bld. 2 Leninsky Ave, Moscow 119071, Russia, Tel: 74959544088, Email: renata_z@inbi.ras.ru

Received: 16-Oct-2017 / Accepted Date: 23-Oct-2017 / Published Date: 30-Oct-2017 DOI: 10.4172/2161-0460.1000394

Abstract

Alzheimer’s disease is a multifaceted, incurable neurologic disorder characterized by cognitive decline and degeneration of brain neurons. The main factors implicated in Alzheimer’s disease including accumulation of misfolded and aggregated proteins (hyperphosphorylated microtubule associated protein referred to as tau and amyloid Aβ), oxidative damage, inflammation, mitochondrial impairments and chronic energy imbalance, chronic endoplasmic reticulum stress, autophagy dysfunction, the abnormality and dysfunction of the mitochondrion-associated endoplasmic reticulum membrane serving as bridges between endoplasmic reticulum and mitochondria and regulating multiple functions such as Ca2+ transfer, energy exchange, lipid synthesis and transports and protein folding, genetic variation in lysosomal genes, metabolomic changes are shortly considered. A special emphasis was placed on mitochondrial fission (fragmentation) is a prominent early event preceding Alzheimer’s disease pathology in transgenic Aβ-animal models, as well as on marked decrease in extracellular amyloid deposition, prevention of the cognitive deficit development and improvement of synaptic parameters after inhibiting abnormalities in mitochondrial dynamics. The important role of the well-characterized Saccharomyces cerevisiae yeast as a valuable eukaryotic model organism in unraveling complex fundamental intracellular mechanisms underlying Alzheimer’s disease is highlighted. The benefits of applying a new model organism the yeast Yarrowia lipolytica, an obligate aerobe with the respiratory metabolism closely resembling that of mammalian cells, amenable to both classical and molecular genetic techniques, having a long history of use as a producer of heterological proteins, possessing an ability to change its morphology (from yeast-like to true mycelium) in response to environmental conditions as an useful alternative in deciphering a role of mitochondrial dynamics and distribution in an yeast model of Alzheimer’s disease are suggested.

Keywords: Alzheimer’s disease; Amyloid-β peptide; Tau protein; Mitochondrial dysfunction; Oxidative stress; Yeast

Abbreviations

Aβ: Amyloid-β Peptide; AD: Alzheimer’s Disease; APP: Amyloid Precursor Protein; ATG: Autophagy-related Protein; Drp1p: Dynamin-Related Protein 1; ER: Endoplasmic Reticulum; ERK: Extracellular signal–Regulated Kinase; HIF: Hypoxia-Inducible Factor; MAM: Mitochondrion-Associated Endoplasmic Reticulum Membrane; MAP: Microtubule Associated Protein; MAPK: Mitogen- Activated Protein Kinase; Mdivi-1:3-(2,4-Dichloro-5-methoxyphenyl)- 2,3-dihydro-2-thioxo-4(1H)-quinazolinone; mtDNA: Mitochondrial DNA; NF-κB: Nuclear Factor Kappa B; PSEN1: Presenilin 1; PSEN2: Presenilin 2; ROS: Reactive Oxygen Species; STAT3: Signal Transducer and Activator of Transcription 3; TNFα: Tumor Necrosis Factor α

Introduction

Alzheimer’s disease (AD) is a multifaceted, incurable neurologic disorder characterized by cognitive decline and degeneration of brain neurons. AD is the most common form of dementia and one of the most important causes of morbidity and mortality among the aging population presently affecting more than 45 million worldwide [1,2] and its prevalence is increasing with the demographic trend of increasing elderly populations in industrialized countries [3,4].

Since AD was first described in 1907, [5] many attempts have been made to reveal its main cause. Nowadays, two forms of the disease are known, and while the very rare hereditary early-onset form of AD is clearly caused by mutations encoding amyloid precursor protein (APP) and presenilin 1 and 2 (PSEN1, PSEN2) [6], fundamental pathogenic mechanisms as well as most hereditary contributions to the predominant age-related sporadic form of AD remain largely unknown.

Hallmarks Of AD

Accumulation of misfolded and aggregated proteins in AD, loss of synapses, neuronal death

Both hereditary and sporadic forms of AD forms share similar sets of neuropathological manifestations, including accumulation of misfolded and aggregated proteins, the extracellular deposition of senile of plaques, followed by the intracellular neurofibrillary tangles, consisting mainly of aggregates of hyperphosphorylated microtubule associated protein (MAP) referred to as tau [7,8]. MAP tau is a key protein in stabilizing the microtubule architecture that regulates neuron morphology and synaptic strength. In the course of AD hyperphosphorylated tau gets truncated by proteolytic cleavage, is subject to O-glycosylation, sumoylation, ubiquitinylation, acetylation and some other modifications [3,4]. When MAP tau is degraded in tauopathic disorders, neuron dysfunction results [9]. Senile plaques are deposits of the amyloid-β peptide (Aβ) produced by the sequential cleavage of β- and γ-secretase at the C terminus of APP. This peptide has an extraordinary ability to undergo conformational changes and is highly amyloidogenic. More than 20 mutations in APP have been linked to familial AD that have altered APP processing with respect to enhanced Aβ generation or aggregation). It is now generally accepted that a progressive accumulation of Aβ aggregates eventually triggers a cascade of cellular changes, including mitochondrial oxidative damage, the hyperphosporylation of tau, synaptic failure and inflammation [10]. This is associated with the loss of synapses and then neuronal death, initially in focal areas including the entorhinal cortex and hippocampus, and ultimately more broadly in the cortex [11]. The loss of synapses in the affected brain regions correlates best with cognitive impairment in AD patients and has been considered as the early mechanism that precedes neuronal loss.

However, the amyloid cascade hypothesis, postulating the key role of Aβ in AD development does not fully explain all of the molecular abnormalities in AD [12]. Evidence is presented suggesting amyloid/ oligomers as necessary but insufficient causes of the dementia and that, for dementia to develop, additional cofactors are required [13]. Those cofactors include several subcellular processes including oxidative damage [10,14-23], recruitment of peripheral immune cells and excessive production of pro-inflammatory mediators [10,24-29], mitochondrial impairments and chronic energy imbalance [12,20,30-47], chronic endoplasmic reticulum (ER) stress [48] and autophagy dysfunction [22,49-54], the abnormality and dysfunction of MAM (the mitochondrion-associated endoplasmic reticulum membrane serving as bridges between ER and mitochondria and regulating multiple functions such as Ca2+ transfer, energy exchange, lipid synthesis and transports and protein folding [55-57], genetic variation in lysosomal genes [58].

Mitochondrial dysfunction in AD

Recently, multiple lines of evidence have stated major roles for the accumulation of mitochondrial dysfunctions, coupled with increased reactive oxygen species (ROS) generation, defects in mitochondrial biogenesis and transport/distribution, aberrant cell cycle re-entry in neurons and mitophagy in sporadic AD etiopathogenesis (so called mitochondrial cascade hypothesis) [12].

Neurons, post-mitotic and excitable cells have high energy requirements to maintain the resting potential through ion pumps, releasing neurotransmitters during synaptic transmissions to communicate with other neurons, and transporting organelles by consuming ATP [30,42]. They rely almost exclusively on the mitochondrial oxidative phosphorylation system to fulfill their energy needs. In addition, neurons are exceedingly compartmentalized, comprising structures like: cell body, axon, dendrites and even more specific compartments that are the synapses, which makes a proper mitochondrial distribution pivotal to sustaining the energy requirement at specific locations within the different neuronal compartments [31,35,36]. The crucial role of mitochondria in supporting synaptic function and the concomitant occurrence of impaired mitochondrial energy production, deregulated mitochondrial calcium handling, excess of mitochondrial ROS generation and release with mediating synaptic transmission deregulation in AD seem to lend the credibility to the hypothesis that mitochondrial defects underlie synaptic failure in AD [32-34,37,41,43,47].

A number of reports suggest the involvement of mitochondrial alterations through intracellular accumulation of oligomeric Aβ. One of the possible mediators for Aβ-impaired mitochondrial function is thought to be the nuclear factor kappa B (NF-κB) signaling pathway, playing important roles in brain inflammation and antioxidant defense, as well as in the regulation of mitochondrial function; studies have confirmed altered NF-κB signaling in AD brain [51]. The mitochondrial alterations include increased ROS production, mitochondrial DNA (mtDNA) depletion, decreased oxidative phosphorylation and ATP production, membrane depolarization, reduced number of mitochondria etc. All these defects cumulatively caused neural toxicity and alterations in cellular energy homeostasis, a significant reduction in neuronal viability [44,45].

However, dysfunctional mitochondria located in synapses can trigger synaptotoxicity through multifaceted mechanisms and that it is not the susceptibility of mitochondria to Aβ [59]. Complementary, in some cases beneficial effects of some agents on survival and cognitive performance were independent of Aβ levels and amyloid plaque deposition, but were associated with improved brain mitochondrial respiration, a reversal of mitochondrial complex I dysfunction, restored ATP production and reduced ROS levels [38].

Many investigators have suggested that epigenetic changes in the copy numbers of mtDNA and mtDNA mutations might be involved in AD pathogenesis [60-65]. However, an analysis of the literature reveals the existence of inconsistent findings and methodological shortcomings affecting a large proportion of mtDNA association studies on AD [66-69].

Unlike static organelles, mitochondria in various eukaryotes change size and shape by undergoing fission and fusion, processes that are orchestrated by the cellular machinery comprised of dynamin-related proteins [70]. It is argued that this kind of organellar dynamics has the power to restore the function of impaired organelles by content mixing with intact organelles.

Excessive mitochondrial fission (fragmentation) is a prominent early event, contributing to mitochondrial dysfunction, synaptic failure, and neuronal cell death in the progression of AD [40,71,72]. Moreover, mitochondria fragmentation, like a mitochondrial bioenergetic deficit, is an early feature preceding AD pathology in APP transgenic animal models [40] and mouse model CRND8 [73].

Treatment by Mdivi-1, a mitochondrial fission inhibitor, rescued the mitochondrial fragmentation and distribution deficits and improve mitochondrial function in the Aβ-treated [72] and CRND8 [73] neurons both in vitro and in vivo. Importantly, Mdivi-1 treatment markedly decreased extracellular amyloid deposition, prevented the development of cognitive deficits in Y-maze test and improved synaptic parameters, supporting the notion that abnormal mitochondrial dynamics plays an early and causal role in mitochondrial dysfunction and ADrelated pathological and cognitive impairments in vivo [72,73]. These results suggest that neuropathology and combined cognitive decline can be attributed to hyperactivation of Drp1p (responsible for mitochondrial fragmentation) in the pathogenesis of AD and that inhibiting excessive Drp1p-mediated mitochondrial fission may be a new efficient therapeutic strategy for AD. However, according to other data [46], Mdivi-1 works better in prevention than treatment in AD neurons.

Mitochondria in neurons challenged with extracellular Aβ and neurons expressing APP show decreased motility and density in axons. Similarly, tau, especially hyperphosphorylated tau, disrupts mitochondrial transport in neuronal cells. An Aβ- or APP- induced mitochondrial trafficking deficit could be alleviated by inhibiting mitochondrial fragmentation, indicating the impairment of mitochondrial movement possibly downstream of mitochondrial fragmentation [40].

Oxidative stress in AD

Oxidative stress reflects an imbalance between the generation and clearance of ROS [14]. ROS is formed as a natural byproduct of metabolism and has important roles in cell signaling and homeostasis. However, excessive production of ROS, largely derived from mitochondrial dysfunction [16], can induce significant damage to cell components, including DNA, proteins and lipids [15] and disturb multiple cellular signaling, including NF-κB, HIF and STAT3 pathways, leading to expression of proteins that control inflammation, cellular transformation, survival and metastasis [74-76]. Oxidative stress has been shown to increase with age in the brain, where oxidative damage is a major contributor to functional decline [10]. Moreover, oxidative stress is also the major cause of glial inflammation and apoptosis. All these findings suggest a critical role for oxidative stress in promoting AD and highlight the potential for antioxidants as potential drugs for combating AD [17-23].

Inflammation in AD

Inflammation is a complex and dynamic process, and during the course of AD, it probably has protective and deleterious effects in different phases [29]. The initial accumulation of Aβ triggers glial cells, such as microglia and astrocytes, the resident immune cells of the central nervous system, which subsequently activates immune reactions. Microglia can identify and bind to Aβ oligomers and fibrils through receptors present on the cell surface; microglia activation reduces Aβ deposits by increasing its phagocytosis. Aβ, in turn, is able to activate the, NF-κB pathway which is a central signaling pathway for cytokine production [24]. When astrocytes are stimulated by proinflammatory cytokines such as IL-1 and IL-6, they become activated (reactive astrocytes) and promote inflammation through the secretion of cytokines such as tumor necrosis factor α (TNF-α) and IL-6. In addition to having a direct cytotoxic effect on the adjacent neurons, these cytokines result in a decrease in scavenger receptors and Aβ- degrading enzymes in the microglia, canceling their neuroprotective role as the disease progresses. The ongoing production and release of pro-inflammatory cytokines (such as TNFα, interleukin-1, interleukin-12, and interleukin-23), prostaglandin E2, NO, ROS, and matrix metalloproteinases leads to a chronic inflammatory state and microglial dysfunction that hinders Aβ clearance [27]. Chemokines such as TNFα may enhance APP and Aβ peptide production [26]. Additionally, Aβ may directly bind to the surface of microglial cells for the activation of MAPK/ERK pathway and induce pro-inflammatory genes including cytokines and chemokines [25], thus leading to a downward spiral of chronic inflammation and causing direct neuronal cell damage and furthering the pathogenesis of AD.

ER stress in AD pathology

Mounting evidence suggests that ER stress is involved in the pathology of AD. The ER is an organelle that functions to facilitate protein folding. However, exposure to stress results in loss of function and causes ER stress. Intriguingly, crosstalk between ER stress and immune function has been suggested [48]. However, the mechanisms linking the progression of AD with ER and immunological stress are still not clear.

Autophagy in AD genesis

Autophagy (from the Greek meaning “selfeating”) is a key homeostatic evolutionarily conserved catabolic process involved in the lysosomal degradation of dysfunctional or unnecessary cellular components (e.g., organelles and proteins [77]. Mechanistically, the cellular components (misfolded proteins (aggrephagy), overloaded peroxisomes (pexophagy), pathogenic organisms (xenophagy) and dysfunctional mitochondria (mitophagy)) are engulfed by autophagosomes which then move towards and fuse with lysosomes and are then degraded [53]. A complicated series of signaling cascades and autophagy-related proteins (ATG) contribute to the completion of autophagy [50].

Recent studies have revealed the protective role of autophagy in neurophysiology, particularly, in AD). Autopaghy negatively regulates inflammation. Increased activity of the autophagy pathway leads to increased degradation of the tau protein and hence reduced intracellular tau aggregation [49], suggesting that the aberrant accumulation of tau proteins may, at least in part, be due to impaired autophagy inside neurons [54]. Another major protective role of autophagy is the elimination of abnormal mitochondria, a source of oxidative stress. The molecular interaction between autophagy and Aβ remains controversial [52] and more in-depth investigations are still needed to clarify the functional role of autophagy on pathological alteration of AD.

Metabolic changes associated with AD

Metabolomic studies have also shown that a range of fundamental changes occur during AD progression. Mounting evidence suggests a link between diabetes, obesity, non-alcoholic fatty liver disease and the progression of AD [78].

Metabolic changes observed in AD patients and ADP models include glucose breakdown and pyruvate oxidation [79], impairment of protein synthesis in early-stage AD [80], increased levels of some amino acids, serotonin, catecholamine and Krebs cycle metabolites [79,81,82], alterations in purine metabolic pathways [79,81], imbalanced cholesterol [83] and sphingolipid [84] homeostasis, large networkwide disruptions in ceramide and phosphoinositide biosynthesis and signaling [85], disruptions in the cellular systems for handling (uptake, intracellular transport, protein loading and storage) transition metals [86,87], dysregulated one-carbon metabolism [88] and some others.

Models for AD

Mouse models for AD

Within the scope of mini-review, we only recall that mouse models, which feature highly genetic kinship with the human genome, have been widely regarded as a suitable tool for AD researches. However, despite many scientific achievements [52], due to the limits of shorter lifespan of mouse and complicated cause of AD, AD-like mouse models do not fully recapitulate human AD pathology, thus there is a need to generation more robust models closely resembling the human pathophysiology of AD.

Yeast models for AD

On the other hand, the growing need to better understand the molecular basis of AD with its diversity of symptoms and sophisticated cross-talk of cofactors has led to the development of simple eukaryotic models amenable for mechanistic studies. In recent years, the field of neurodegeneration, AD particularly, has derived significant benefit from the use of the simple and well-characterized eukaryote Saccharomyces cerevisiae [89]. Yeast cells possess most of the same fundamental cellular machinery as neurons in the brain. Moreover, numerous processes and mechanisms such as cell signaling pathways that regulate metabolism, cell growth and division, organelle function, cellular homeostasis and stress responses were first identified in yeasts and then shown to be conserved in higher eukaryotes. The high degree of conservation between yeast and higher eukaryotes is one of the reasons why yeast cells are so reliable as biological model for age-related diseases [90,91]. Despite nearly a billion years of evolutionary divergence, recent estimates showed that a fifth of yeast genes have human disease orthologs lending support to functional discovery investigations using this model [92]. Moreover, thanks to amenability of S. cerevisiae to both classical and advanced molecular genetic techniques, to relatively simple, cheap and quick genetic and environmental manipulations, to the large knowledge base and data collections, high-throughput screening technologies and functional genomics that are not possible in humans [93-95], this organism has become a valuable and prevalent eukaryotic model organism to unravel complex and fundamental intracellular mechanisms underlying neurodegeneration [96-104].

Humanized yeasts are also utilized in high-throughput screening of genes that affect the toxicity of heterologously expressed human proteins, for large-scale chemical screens aiming at the discovery of novel compounds delaying aging or protecting against human agerelated diseases.

Several excellent reviews have dealt with specific aspects of AD, including Aβ toxicity, using this model ([3,4,7,102,105,106] and references therein).

Improved yeast models for AD

However, the fermentation-oriented yeast S. cerevisiae (selected for thousands of years for its capacity for alcoholic fermentation) with less abundant mitochondria is hardly a bioenergetic equivalent of high-energy demanding neurons relying almost exclusively on mitochondrial oxidative phosphorylation. S. cerevisiae is not the best model organism for studying fragmentation of mitochondria, as they contain small-sized, poorly structured mitochondria. In these respects, Yarrowia lipolytica, a non-toxic ascomycetous yeast species having a haploid genome and sexual life cycle and amenable to both classical and molecular genetic techniques [107], an obligate aerobe with the respiratory metabolism closely resembling that of mammalian cells [108,109], vigorously growing on a variety of simple, well defined and inexpensive media [110], having a long history of use as a producer of heterological proteins [111], possessing an ability to change its morphology (from yeast-like to true mycelium) in response to environmental conditions [112,113], may be a useful alternative in deciphering a role of mitochondrial dynamics and distribution in an yeast model of AD.

Limitations of yeast models for AD

However, it should be noted that although yeast offers many advantages for mechanistic dissection of neurotoxic disorders and has contributed greatly to the understanding of the molecular underpinnings of human diseases [89,106], there are some natural limitations; unicellular organisms such as yeast fails as a model to study the multicellularity and cell-cell interactions, particularly important in the neuronal cross-talk that is of major importance to neurodegeneration [114]. Yeasts lack neuron-specific morphological structures, such as dendrites, axons and synapses. Consequently, the underlying neuron-specific molecular inventories are missing. Therefore, age-related and disease-associated processes uncovered in yeasts must be carefully extrapolated to human aging and diseases. Ultimately, these findings must be validated in neuronal model systems and more complex eukaryotic models.

Conclusion

AD is a complex, multifaceted neurologic disorder characterized by cognitive decline and degeneration of brain neurons, arising from interplay of many factors including accumulation of misfolded and aggregated proteins, mitochondrial impairments, oxidative damage, inflammation, endoplasmic reticulum stress, autophagy dysfunction, the abnormalities in Ca2+ homeostasis, transfer and energy exchange, protein folding, genetic variation in lysosomal genes, metabolic changes and others. Despite tremendous efforts in elucidating the molecular and cellular players involved in AD pathology, to date, there is no treatment that could prevent or cure this disease. Current treatments are only useful in slowing down the progression of AD and helping patients manage some of their behavioral and cognitive symptoms instead of reversing the ultimate consequence [115]. These unsatisfactory effects force people to pay attention on more targeted and etiology-oriented strategies [116]. Due to the notion that age-related oxidative stress has been acknowledged as one of the major risk factors of AD pathogenesis and the early manifestation of AD [18], anti-aging drugs (antioxidants) have become the prevailing therapeutic strategy against AD. In this respect, yeasts, relatively simple unicellular organisms, vigorously growing on simple and inexpensive media may be exceptionally promising models for searching for and treating the newly synthesized effective antioxidants, especially mitochondria-targeted antioxidants [117-119]. Further research exploring these data and other finding obtained from analysis of other models will help put together the pieces of the puzzle to create a unifying theory of this highly complex AD process.

Financial Support

This manuscript was supported by the Russian Foundation for Basic Research (grants 16-04-01040 and 16-34-01732).

References

  1. Frej AD, Otto GP, Williams RSB (2017) Tipping the scales: Lessons from simple model systems on inositol imbalance in neurological disorders. Eur J Cell Biol 96: 154-163.
  2. Morales I, Cerda-Troncoso C, Andrade V, Maccioni RB (2017) The natural product curcumin as a potential coadjuvant in Alzheimer's treatment. J Alzheimers Dis 60: 451-460.
  3. Heinisch JJ, Brandt R (2016) Signaling pathways and posttranslational modifications of tau in Alzheimer's disease: The humanization of yeast cells. Microb Cell 3: 135-146.
  4. Zhang X, Wang WA, Jiang LX, Liu HY, Zhang BZ, et al. (2017) Downregulation of RBO-PI4KIIIα facilitates Aβ42 secretion and ameliorates neural deficits in Aβ42-Expressing drosophila. J Neurosci 37: 4928-4941.
  5. Alzheimer A (1907) Uber eine eigenartige erkrankung der hirnrinde. Allg Zeitschr Psychiatrie 64: 146-148.
  6. Remes AM, Finnila S, Mononen H, Tuominen H, Takalo R, et al. (2004) Hereditary dementia with intracerebral hemorrhages and cerebral amyloid angiopathy. Neurology 63: 234-240.
  7. Verduyckt M, Vignaud H, Bynens T, Van den Brande J, Franssens V, et al. (2016) Yeast as a model for Alzheimer's disease: Latest studies and advanced strategies. Methods Mol Biol 1303: 197-215.
  8. França MB, Lima KC, Eleutherio EC (2017) Oxidative stress and amyloid toxicity: Insights from yeast. J Cell Biochem 118: 1442-1452.
  9. Kurian P, Obisesan TO, Craddock TJA (2017) Oxidative species-induced excitonic transport in tubulin aromatic networks: Potential implications for neurodegenerative disease. J Photochem Photobiol B 175: 109-124.
  10. Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, et al. (2006) Mitochondria are a direct site of A beta accumulation in Alzheimer's disease neurons: Implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet 15: 1437-1449.
  11. Huang Y, Mucke L (2012) Alzheimer mechanisms and therapeutic strategies. Cell 148: 1204-1222.
  12. Kozlov S, Afonin A, Evsyukov I, Bondarenko A (2017) Alzheimer's disease: As it was in the beginning. Rev Neurosci.
  13. Fessel J (2017) Amyloid is essential but insufficient for Alzheimer causation: Addition of subcellular cofactors is required for Dementia. Int J Geriatr Psychiatry.
  14. Mittler R (2002) Oxidative stress, antioxidants and stress tolerance. Trends Plant Sci 7: 405-410.
  15. Cooke MS, Evans MD, Dizdaroglu M, Lunec J (2003) Oxidative DNA damage: Mechanisms, mutation and disease. FASEB J 17: 1195-1214.
  16. Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443: 787-795.
  17. Chmatalova Z, Vyhnalek M, Laczo J, Hort J, Pospisilova R, et al. (2017) Relation of plasma selenium and lipid peroxidation end products in patients with Alzheimer's disease. Physiol Res.
  18. Guo L, Tian J, Du H (2017) Mitochondrial dysfunction and synaptic transmission failure in Alzheimer's disease. J Alzheimers Dis 57: 1071-1086.
  19. Guo J, Cheng J, North BJ, Wei W (2017) Functional analyses of major cancer-related signaling pathways in Alzheimer's disease etiology. Biochim Biophys Acta 1868: 341-358.
  20. Islam MT (2017) Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol Res 39: 73-82.
  21. Kolosova NG, Tyumentsev MA, Muraleva NA, Kiseleva E, Vitovtov AO, et al. (2017) Antioxidant SkQ1 alleviates signs of Alzheimer's disease-like pathology in old OXYS rats by reversing mitochondrial deterioration. Curr Alzheimer Res.
  22. Murphy KE, Park JJ (2017) Can co-activation of Nrf2 and neurotrophic signaling pathway slow Alzheimer's disease? Int J Mol Sci 18: E1168.
  23. Vergallo A, Giampietri L, Baldacci F, Volpi L, Chico L, et al. (2017) Oxidative stress assessment in Alzheimer's disease: A clinic setting study. Am J Alzheimers Dis Other Demen.
  24. Akama KT, Albanese C, Pestell RG, Van Eldik LJ (1998) Amyloid beta-peptide stimulates nitric oxide production in astrocytes through an NFkappaB-dependent mechanism. Proc Natl Acad Sci U S A 95: 5795-5800.
  25. Bell KA, O'Riordan KJ, Sweatt JD, Dineley KT (2004) MAPK recruitment by beta-amyloid in organotypic hippocampal slice cultures depends on physical state and exposure time. J Neurochem 91: 349-361.
  26. Valerio A, Boroni F, Benarese M, Sarnico I, Ghisi V, et al. (2006) NF-kappaB pathway: A target for preventing beta-amyloid (Abeta)-induced neuronal damage and Abeta42 production. Eur J Neurosci 23: 1711-1720.
  27. Heppner FL, Ransohoff RM, Becher B (2015) Immune attack: The role of inflammation in Alzheimer disease. Nat Rev Neurosci 16: 358-372.
  28. Rojas-Gutierrez E, Muñoz-Arenas G, Treviño S, Espinosa B, Chavez R, et al. (2017) Alzheimer's disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse.
  29. Takada LT (2017) Innate immunity and inflammation in Alzheimer´s disease pathogenesis. Arq Neuropsiquiatr 75: 607-608.
  30. Harris JJ, Jolivet R, Attwell D (2012) Synaptic energy use and supply. Neuron 75: 762-777.
  31. Obashi K, Okabe S (2013) Regulation of mitochondrial dynamics and distribution by synapse position and neuronal activity in the axon. Eur J Neurosci 38: 2350-2363.
  32. Silva DF, Selfridge JE, Lu J, E Lezi, Roy N, et al. (2013) Bioenergetic flux, mitochondrial mass and mitochondrial morphology dynamics in AD and MCI cybrid cell lines. Hum Mol Genet 22: 3931-3946.
  33. Reddy PH (2014) Inhibitors of mitochondrial fission as a therapeutic strategy for diseases with oxidative stress and mitochondrial dysfunction. J Alzheimers Dis 40: 245-256.
  34. Cabezas-Opazo FA, Vergara-Pulgar K, Pérez MJ, Jara C, Osorio-Fuentealba C, et al. (2015) Mitochondrial dysfunction contributes to the pathogenesis of Alzheimer's disease. Oxid Med Cell Longev 2015: 509654
  35. Lin MY, Sheng ZH (2015) Regulation of mitochondrial transport in neurons. Exp Cell Res 334: 35-44.
  36. Pernas L, Scorrano L (2016) Mito-morphosis: Mitochondrial fusion, fission and cristae remodeling as key mediators of cellular function. Annu Rev Physiol 78: 505-531.
  37. Swerdlow RH (2016) Bioenergetics and metabolism: A bench to bedside perspective. J Neurochem 139: 126-135.
  38. Derungs R, Camici GG, Spescha RD, Welt T, Tackenberg C, et al. (2017) Genetic ablation of the p66Shc adaptor protein reverses cognitive deficits and improves mitochondrial function in an APP transgenic mouse model of Alzheimer's disease. Mol Psychiatry 22: 605-614.
  39. Gajewski M, Rzodkiewicz P, Maśliński S (2017) The human body as an energetic hybrid? New perspectives for chronic disease treatment? Reumatologia 55: 94-99.
  40. Gao J, Wang L, Liu J, Xie F, Su B, et al. (2017) Abnormalities of mitochondrial dynamics in neurodegenerative diseases. Antioxidants (Basel) 6.
  41. Guo XD, Sun GL, Zhou TT, Wang YY, Xu X, et al. (2017) LX2343 alleviates cognitive impairments in AD model rats by inhibiting oxidative stress-induced neuronal apoptosis and tauopathy. Acta Pharmacol Sin 38: 1104-1119.
  42. Kim DI, Lee KH, Oh JY, Kim JS, Han HJ, et al. (2017) Relationship between β-amyloid and mitochondrial dynamics. Cell Mol Neurobiol 37: 955-968.
  43. Naia L, Ferreira IL, Ferreiro E, Rego AC (2017) Mitochondrial Ca2+ handling in Huntington's and Alzheimer's diseases - Role of ER-mitochondria crosstalk. Biochem Biophys Res Commun 483: 1069-1077.
  44. Onyango IG, Khan SM, Bennett JP (2017) Mitochondria in the pathophysiology of Alzheimer's and Parkinson's diseases. Front Biosci (Landmark Ed) 22: 854-872.
  45. Parmar HS, Houdek Z, Pesta M, Vaclava C, Dvorak P, et al. (2017) Protective effect of aspirin against oligomeric Aβ42 induced mitochondrial alterations and neurotoxicity in differentiated EC P19 neuronal cells. Curr Alzheimer Res 14: 810-819.
  46. Swerdlow RH, Koppel S, Weidling I, Hayley C, Ji Y, et al. (2017) Mitochondria, cybrids, aging and Alzheimer's disease. Prog Mol Biol Transl Sci 146: 259-302.
  47. Völgyi K, Háden K, Kis V, Gulyássy P (2017) Mitochondrial proteome changes correlating with β-amyloid accumulation. Mol Neurobiol 54: 2060-2078.
  48. Hosoi T, Ozawa K (2012) Molecular approaches to the treatment, prophylaxis and diagnosis of Alzheimer's disease: Endoplasmic reticulum stress and immunological stress in pathogenesis of Alzheimer's disease. J Pharmacol Sci 118: 319-324.
  49. Zare-Shahabadi A, Masliah E, Johnson GV, Rezaei N (2015) Autophagy in Alzheimer's disease. Rev Neurosci 26: 385-395.
  50. Cheng J, Liao Y, Xiao L, Wu R, Zhao S, et al. (2017) Autophagy regulates MAVS signalling activation in a phosphorylation-dependent manner in microglia. Cell Death Differ 24: 276-287.
  51. Djordjevic J, Thomson E, Chowdhury SR, Snow WM, Perez C, et al. (2017) Brain region- and sex-specific alterations in mitochondrial function and NF-κB signaling in the TgCRND8 mouse model of Alzheimer's disease. Neuroscience 361: 81-92.
  52. Gou H, Zhao M, Xu H, Yuan J, He W, et al. (2017) CSFV induced mitochondrial fission and mitophagy to inhibit apoptosis. Oncotarget 8: 39382-39400.
  53. Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, et al. (2017) Autophagy and neurodegeneration: Pathogenic mechanisms and therapeutic opportunities. Neuron 93: 1015-1034.
  54. Li Q, Liu Y, Sun M (2017) Autophagy and Alzheimer's disease. Cell Mol Neurobiol 37: 377-388.
  55. Erpapazoglou Z, Mouton-Liger F, Corti O (2017) From dysfunctional endoplasmic reticulum-mitochondria coupling to neurodegeneration. Neurochem Int 109: 171-183.
  56. Liu Y, Zhu X (2017) Endoplasmic reticulum-mitochondria tethering in neurodegenerative diseases. Transl Neurodegener 6: 21.
  57. Weng TY, Tsai SA, Su TP (2017) Roles of sigma-1 receptors on mitochondrial functions relevant to neurodegenerative diseases. J Biomed Sci 24: 74.
  58. Whyte LS, Lau AA, Hemsley KM, Hopwood JJ (2017) Endo-lysosomal and autophagic dysfunction: A driving factor in Alzheimer's disease? J Neurochem 140: 703-717.
  59. Amorim JA, Canas PM, Tomé AR, Rolo AP (2017) Mitochondria in excitatory and inhibitory synapses have similar susceptibility to amyloid-β peptides modeling Alzheimer's disease. J Alzheimers Dis 60: 525-536.
  60. Duran GP, Martinez-Aguayo A, Poggi H, Lagos M, Gutierrez D, et al. (2012) Large mitochondrial DNA deletion in an infant with addison disease. JIMD Rep 3: 5-9.
  61. Chen Y, Liu C, Parker WD, Chen H, Beach TG, et al. (2016) Mitochondrial DNA rearrangement spectrum in brain tissue of Alzheimer's disease: Analysis of 13 Cases. PLoS One 11: e0154582.
  62. Delbarba A, Abate G, Prandelli C, Marziano M, Buizza L, et al. (2016) Mitochondrial alterations in peripheral mononuclear blood cells from Alzheimer's disease and mild cognitive impairment patients. Oxid Med Cell Longev 2016: 5923938.
  63. Hoekstra JG, Hipp MJ, Montine TJ, Kennedy SR (2016) Mitochondrial DNA mutations increase in early stage Alzheimer disease and are inconsistent with oxidative damage. Ann Neurol 80: 301-306.
  64. Reznik E, Miller ML, Şenbabaoğlu Y, Riaz N, Sarungbam J, et al. (2016) Mitochondrial DNA copy number variation across human cancers. Elife 5.
  65. Stoccoro A, Siciliano G, Migliore L, Coppedè F (2017) Decreased methylation of the mitochondrial D-loop region in late-onset Alzheimer's disease. J Alzheimers Dis 59: 559-564.
  66. Fachal L, Mosquera-Miguel A, Pastor P, Ortega-Cubero S, Lorenzo E, et al. (2015) No evidence of association between common European mitochondrial DNA variants in Alzheimer, Parkinson and migraine in the Spanish population. Am J Med Genet B Neuropsychiatr Genet 168B: 54-65.
  67. Mastroeni D, Khdour OM, Delvaux E, Nolz J, Olsen G, et al. (2017) Nuclear but not mitochondrial-encoded oxidative phosphorylation genes are altered in aging, mild cognitive impairment and Alzheimer's disease. Alzheimers Dement 13: 510-519.
  68. Pienaar IS, Howell N, Elson JL (2017) MutPred mutational load analysis shows mildly deleterious mitochondrial DNA variants are not more prevalent in Alzheimer's patients, but may be under-represented in healthy older individuals. Mitochondrion 34: 141-146.
  69. Wei W, Keogh MJ, Wilson I, Coxhead J, Ryanet S, et al. (2017) Mitochondrial DNA point mutations and relative copy number in 1363 disease and control human brains. Acta Neuropathol Commun 5: 13.
  70. Chen H, Chan DC (2005) Emerging functions of mammalian mitochondrial fusion and fission. Hum Mol Genet 14: R283-289.
  71. Reddy PH, Manczak M, Yin X (2017) Mitochondria-division inhibitor 1 protects against amyloid- β induced mitochondrial fragmentation and synaptic damage in Alzheimer's disease. J Alzheimers Dis 58: 147-162.
  72. Baek SH, Park SJ, Jeong JI, Kim J, Han J, et al. (2017) Inhibition of Drp1 ameliorates synaptic depression, abeta deposition and cognitive impairment in an Alzheimer's Disease Model. J Neurosci 37: 5099-5110.
  73. Wang W, Yin J, Ma X, Zhao F (2017) Inhibition of mitochondrial fragmentation protects against Alzheimer's disease in rodent model. Hum Mol Genet.
  74. Hancock JT, Desikan R, Neill SJ (2001) Role of reactive oxygen species in cell signalling pathways. Biochem Soc Trans 29: 345-349.
  75. Qutub AA, Popel AS (2008) Reactive oxygen species regulate hypoxia-inducible factor 1alpha differentially in Cancer and Ischemia. Mol Cell Biol 28: 5106-5119.
  76. Morgan MJ, Liu ZG (2011) Crosstalk of reactive oxygen species and NF- κB signaling. Cell Res 21: 103-115.
  77. Alirezaei M, Kemball CC, Whitton JL (2011) Autophagy, inflammation and neurodegenerative disease. Eur J Neurosci 33: 197-204.
  78. Jha SK, Jha NK, Kumar D, Ambasta RK, Kumar P (2017) Linking mitochondrial dysfunction, metabolic syndrome and stress signaling in Neurodegeneration. Biochim Biophys Acta 1863: 1132-1146.
  79. Hoyer S (1990) Brain glucose and energy metabolism during normal aging. Aging (Milano) 2: 245-258.
  80. Wang H, Hong X, Li S, Wang Y (2017) Oxygen supplementation improves protein milieu supportive of protein synthesis and antioxidant function in the cortex of Alzheimer's disease model mice-a quantitative proteomic study. J Mol Neurosci.
  81. Kaddurah-Daouk R, Zhu H, Sharma S, Bogdanov M, Rozen SG, et al. (2013) Alterations in metabolic pathways and networks in Alzheimer's disease. Transl Psychiatry 3: e244.
  82. Frej AD, Clark J, Le Roy CI, Lilla S, Thomason PA, et al. (2016) The inositol-3-phosphate synthase biosynthetic enzyme has distinct catalytic and metabolic roles. Mol Cell Biol 36: 1464-1479.
  83. Korber M, Klein I, Daum G (2017) Steryl ester synthesis, storage and hydrolysis: A contribution to sterol homeostasis. Biochim Biophys Acta 1862: 1534-1545.
  84. Spincemaille P, Cammue BP, Thevissen K (2014) Sphingolipids and mitochondrial function, lessons learned from yeast. Microb Cell 1: 210-224.
  85. Kennedy MA, Gable K, Niewola-Staszkowska K, Abreu S, Johnston A, et al. (2014) A neurotoxic glycerophosphocholine impacts PtdIns-4, 5-bisphosphate and TORC2 signaling by altering ceramide biosynthesis in yeast. PLoS Genet 10: e1004010.
  86. Bleackley MR, Macgillivray RT (2011) Transition metal homeostasis: From yeast to human disease. Biometals 24: 785-809.
  87. Behzadfar L, Abdollahi M, Sabzevari O, Hosseini R, Salimi A, et al. (2017) Potentiating role of copper on spatial memory deficit induced by beta amyloid and evaluation of mitochondrial function markers in the hippocampus of rats. Metallomics 9: 969-980.
  88. Cavallaro RA, Nicolia V, Fiorenza MT, Scarpa S (2017) S-Adenosylmethionine and superoxide dismutase 1 synergistically counteract Alzheimer's disease features progression in TgCRND8 mice. Antioxidants (Basel) 6.
  89. Braun RJ, Buttner S, Ring J, Kroemer G, Madeo F (2010) Nervous yeast: Modeling neurotoxic cell death. Trends Biochem Sci 35: 135-144.
  90. Kaeberlein M (2010) Lessons on longevity from budding yeast. Nature 464: 513-519.
  91. Tenreiro S, Munder MC, Alberti S, Outeiro TF (2013) Harnessing the power of yeast to unravel the molecular basis of neurodegeneration. J Neurochem 127: 438-452.
  92. Peffer S, Cope K, Morano KA (2015) Unraveling protein misfolding diseases using model systems. Future Sci OA 1: FSO41.
  93. Zhang X, Smith DL, Meriin AB, Engemann S, Russel DE, et al. (2005) A potent small molecule inhibits polyglutamine aggregation in Huntington's disease neurons and suppresses neurodegeneration in vivo. Proc Natl Acad Sci U S A 102: 892-897.
  94. Fatouros C, Pir GJ, Biernat J, Koushika SP, Mandelkow E, et al. (2012) Inhibition of tau aggregation in a novel Caenorhabditis elegans model of tauopathy mitigates proteotoxicity. Hum Mol Genet 21: 3587-3603.
  95. Huang Z, Chen K, Zhang J, Li Y, Wang H, et al. (2013) A functional variomics tool for discovering drug-resistance genes and drug targets. Cell Rep 3: 577-585.
  96. Petranovic D, Nielsen J (2008) Can yeast systems biology contribute to the understanding of human disease? Trends Biotechnol 26: 584-590.
  97. Khurana V, Lindquist S (2010) Modelling neurodegeneration in Saccharomyces cerevisiae: Why cook with baker's yeast? Nat Rev Neurosci 11: 436-449.
  98. Munoz AJ, Wanichthanarak K, Meza E, Petranovic D (2012) Systems biology of yeast cell death. FEMS Yeast Res 12: 249-265.
  99. Pimentel C, Batista-Nascimento L, Rodrigues-Pousada C, Menezes RA (2012) Oxidative stress in Alzheimer's and Parkinson's diseases: Insights from the yeast Saccharomyces cerevisiae. Oxid Med Cell Longev 2012: 132146.
  100. Mirisola MG, Braun RJ, Petranovic D (2014) Approaches to study yeast cell aging and death. FEMS Yeast Res 14: 109-118.
  101. Panaretou B, Jones GW (2014) Yeast models for amyloid disease. Essays Biochem 56: 85-97.
  102. Chen X, Petranovic D (2015) Amyloid-β peptide-induced cytotoxicity and mitochondrial dysfunction in yeast. FEMS Yeast Res 15.
  103. Moosavi B, Mousavi B, Macreadie IG (2015) Yeast Model of Amyloid-β and Tau Aggregation in Alzheimer's Disease. J Alzheimers Dis 47: 9-16.
  104. Tardiff DF, Brown LE, Yan X, Trilles R, Jui NT, et al. (2017) Dihydropyrimidine-Thiones and clioquinol synergize to target β-amyloid cellular pathologies through a metal-dependent mechanism. ACS Chem Neurosci 8: 2039-2055.
  105. Shrestha A, Megeney LA (2015) Yeast proteinopathy models: A robust tool for deciphering the basis of neurodegeneration. Microb Cell 2: 458-465.
  106. Oliveira AV, Vilaça R (2017) Exploring the power of yeast to model aging and age-related neurodegenerative disorders. Biogerontology 18: 3-34.
  107. Barth G, Weber H (1985) Improvement of sporulation in the yeast Yarrowia lipolytica. Antonie Van Leeuwenhoek 51: 167-177.
  108. Zvyagilskaya R, Parchomenko O, Abramova N, Allard P, Panaretakis T, et al. (2001) Proton- and sodium-coupled phosphate transport systems and energy status of Yarrowia lipolytica cells grown in acidic and alkaline conditions. J Membr Biol 183: 39-50.
  109. Trendeleva TA, Sukhanova EI, Rogov AG, Zvyagilskaya RA, Seveina II, et al. (2013) Role of charge screening and delocalization for lipophilic cation permeability of model and mitochondrial membranes. Mitochondrion 13: 500-506.
  110. Zvyagilskaya R, Andreishcheva E, Soares MI, Khozin I, Berhe A, et al. (2001) Isolation and characterization of a novel leaf-inhabiting osmo-, salt- and alkali-tolerant Yarrowia lipolytica yeast strain. J Basic Microbiol 41: 289-303.
  111. Sassi H, Delvigne F, Kar T, Nicaud JM, Coq AM, et al. (2016) Deciphering how LIP2 and POX2 promoters can optimally regulate recombinant protein production in the yeast Yarrowia lipolytica. Microb Cell Fact 15: 159.
  112. Ruiz-Herrera J, Sentandreu R (2002) Different effectors of dimorphism in Yarrowia lipolytica. Arch Microbiol 178: 477-483.
  113. Rogov AG, Ovchenkova AP, Goleva TN, Kireev, II, Zvyagilskaya RA (2017) New yeast models for studying mitochondrial morphology as affected by oxidative stress and other factors. Anal Biochem S0003-2697: 30161-30166.
  114. Mohammadi S, Saberidokht B, Subramaniam S, Grama A (2015) Scope and limitations of yeast as a model organism for studying human tissue-specific pathways. BMC Syst Biol 9: 96.
  115. Fakhoury M (2017) Microglia and astrocytes in Alzheimer's disease: Implications for therapy. Curr Neuropharmacol.
  116. Cazarim MS, Moriguti JC, Ogunjimi AT, Pereira LR (2016) Perspectives for treating Alzheimer's disease: A review on promising pharmacological substances. Sao Paulo Med J 134: 342-354.
  117. Chernyak BV, Antonenko YN, Domnina LV, Ivanova OY, Lyamzaev KG, et al. (2013) Novel penetrating cations for targeting mitochondria. Curr Pharm Des 19: 2795-2806.
  118. Severina II, Severin FF, Korshunova GA, Sumbatyan NV, Ilyasova TM, et al. (2013) In search of novel highly active mitochondria-targeted antioxidants: Thymoquinone and its cationic derivatives. FEBS Lett 587: 2018-2024.
  119. Pustovidko AV, Rokitskaya TI, Severina, II, Simonyan RA, Trendeleva TA, et al. (2013) Derivatives of the cationic plant alkaloids berberine and palmatine amplify protonophorous activity of fatty acids in model membranes and mitochondria. Mitochondrion 13: 520-525.

Citation: Goleva T, Rogov A, Zvyagilskaya R (2017) Alzheimer’s Disease: Molecular Hallmarks and Yeast Models. J Alzheimers Dis Parkinsonism 7: 394. DOI: 10.4172/2161-0460.1000394

Copyright: © 2017 Goleva T, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

https://bahigox.fun/ https://bahisjet.fun/ https://bahsegel.fun/ https://betboo.fun/ https://betdoksan.xyz/ https://betebetgiris.xyz/ https://betexper.fun/ https://betgram.fun/ https://betkanyon.fun/ https://betkolik.fun/
Top