ijms-logo

Journal Browser

Journal Browser

Cell Signaling in Neurodegeneration

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Neurobiology".

Deadline for manuscript submissions: closed (31 July 2020) | Viewed by 63257

Special Issue Editor


E-Mail Website
Guest Editor
Achucarro Basque Center for Neuroscience, Barrio de Sarriena, s/n, 48940 Leioa, Spain
Interests: small GTPases; glycogen metabolism; PTMs; neurodegeneration and signaling pathways
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Cellular communication and signaling are keys to coordinate the functions of the different cell types that constitute organisms. These systems control processes such as inter- and intracellular transport, changes in cell morphology, energy consumption, and accumulation, cell differentiation, cell migration, cell proliferation or cell death. These controls are established through gene regulation and the function of small and macromolecules, such as proteins. It is estimated that more than 20% of the genes of the human genome encode proteins are involved in the signal transduction pathways that regulate changes in localization, trafficking, degradation, of molecules or functional intermolecular interactions. Understanding how cells receive and coordinate signals from the environment and/or other cells to finally emit a physiological response is the basis for correcting dysfunctionalities that cause unregulated modifications of these signaling systems and result in multiple pathologies, including neurodegenerative diseases.

The purpose of this Special Issue of IJMS is to explore this paradigm of cell signaling in neurodegeneration in order to understand the molecular mechanisms that lead to these neurodegenerative processes. Therefore, this Special Issue provides an excellent opportunity to cover studies and reports on cell cultures, animal models, revealing new knowledge about the underlying pathophysiology/pathogenesis, or new aspects that could affect positively the progressive development of efficient molecules to fight those neurodegenerative diseases effectively.

Prof. José L Zugaza
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • small GTPases
  • Post-translational modification (PTM)
  • enzyme activity
  • cytoskeleton
  • gene expression
  • epigenetic modifications
  • glia
  • neuron
  • neurodegenration

Published Papers (15 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

3 pages, 202 KiB  
Editorial
Cell Signaling in Neurodegeneration
by José L. Zugaza
Int. J. Mol. Sci. 2021, 22(16), 8978; https://doi.org/10.3390/ijms22168978 - 20 Aug 2021
Cited by 3 | Viewed by 1926
Abstract
Neurodegenerative diseases are characterized by the progressive loss of specific subsets of neurons [...] Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)

Research

Jump to: Editorial, Review

18 pages, 2866 KiB  
Article
The Down-Regulation of Clusterin Expression Enhances the αSynuclein Aggregation Process
by Chiara Lenzi, Ileana Ramazzina, Isabella Russo, Alice Filippini, Saverio Bettuzzi and Federica Rizzi
Int. J. Mol. Sci. 2020, 21(19), 7181; https://doi.org/10.3390/ijms21197181 - 29 Sep 2020
Cited by 16 | Viewed by 2365
Abstract
Parkinson’s Disease (PD) is a progressive neurodegenerative disease characterized by the presence of proteinaceous aggregates of αSynuclein (αSyn) in the dopaminergic neurons. Chaperones are key components of the proteostasis network that are able to counteract αSyn’s aggregation, as well as its toxic effects. [...] Read more.
Parkinson’s Disease (PD) is a progressive neurodegenerative disease characterized by the presence of proteinaceous aggregates of αSynuclein (αSyn) in the dopaminergic neurons. Chaperones are key components of the proteostasis network that are able to counteract αSyn’s aggregation, as well as its toxic effects. Clusterin (CLU), a molecular chaperone, was consistently found to interfere with Aβ aggregation in Alzheimer’s Disease (AD). However, its role in PD pathogenesis has yet to be extensively investigated. In this study, we assessed the involvement of CLU in the αSyn aggregation process by using SH-SY5Y cells stably overexpressing αSyn (SH-Syn). First, we showed that αSyn overexpression caused a strong increase in CLU expression without affecting levels of Hsp27, Hsp70, and Hsp90, which are the chaperones widely recognized to counteract αSyn burden. Then, we demonstrated that αSyn aggregation, induced by proteasome inhibition, determines a strong increase of CLU in insoluble aggregates. Remarkably, we revealed that CLU down-regulation results in an increase of αSyn aggregates in SH-Syn without significantly affecting cell viability and the Unfolded Protein Response (UPR). Furthermore, we demonstrated the direct molecular interaction between CLU and αSyn via a co-immunoprecipitation (co-IP) assay. All together, these findings provide incontrovertible evidence that CLU is an important player in the response orchestrated by the cell to cope with αSyn burden. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

17 pages, 3658 KiB  
Article
STING-Mediated Autophagy Is Protective against H2O2-Induced Cell Death
by Amar Abdullah, Frank Mobilio, Peter J. Crack and Juliet M. Taylor
Int. J. Mol. Sci. 2020, 21(19), 7059; https://doi.org/10.3390/ijms21197059 - 25 Sep 2020
Cited by 8 | Viewed by 3151
Abstract
Stimulator of interferon genes (STING)-mediated type-I interferon signaling is a well characterized instigator of the innate immune response following bacterial or viral infections in the periphery. Emerging evidence has recently linked STING to various neuropathological conditions, however, both protective and deleterious effects of [...] Read more.
Stimulator of interferon genes (STING)-mediated type-I interferon signaling is a well characterized instigator of the innate immune response following bacterial or viral infections in the periphery. Emerging evidence has recently linked STING to various neuropathological conditions, however, both protective and deleterious effects of the pathway have been reported. Elevated oxidative stress, such as neuroinflammation, is a feature of a number of neuropathologies, therefore, this study investigated the role of the STING pathway in cell death induced by elevated oxidative stress. Here, we report that the H2O2-induced activation of the STING pathway is protective against cell death in wildtype (WT) MEFSV40 cells as compared to STING−/− MEF SV40 cells. This protective effect of STING can be attributed, in part, to an increase in autophagy flux with an increased LC3II/I ratio identified in H2O2-treated WT cells as compared to STING−/− cells. STING−/− cells also exhibited impaired autophagic flux as indicated by p62, LC3-II and LAMP2 accumulation following H2O2 treatment, suggestive of an impairment at the autophagosome-lysosomal fusion step. This indicates a previously unrecognized role for STING in maintaining efficient autophagy flux and protecting against H2O2-induced cell death. This finding supports a multifaceted role for the STING pathway in the underlying cellular mechanisms contributing to the pathogenesis of neurological disorders. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

26 pages, 9768 KiB  
Article
Genome Wide Analysis Points towards Subtype-Specific Diseases in Different Genetic Forms of Amyotrophic Lateral Sclerosis
by Banaja P. Dash, Marcel Naumann, Jared Sterneckert and Andreas Hermann
Int. J. Mol. Sci. 2020, 21(18), 6938; https://doi.org/10.3390/ijms21186938 - 21 Sep 2020
Cited by 11 | Viewed by 3547
Abstract
Amyotropic lateral sclerosis (ALS) is a lethally progressive and irreversible neurodegenerative disease marked by apparent death of motor neurons present in the spinal cord, brain stem and motor cortex. While more and more gene mutants being established for genetic ALS, the vast majority [...] Read more.
Amyotropic lateral sclerosis (ALS) is a lethally progressive and irreversible neurodegenerative disease marked by apparent death of motor neurons present in the spinal cord, brain stem and motor cortex. While more and more gene mutants being established for genetic ALS, the vast majority suffer from sporadic ALS (>90%). It has been challenging, thus, to model sporadic ALS which is one reason why the underlying pathophysiology remains elusive and has stalled the development of therapeutic strategies of this progressive motor neuron disease. To further unravel these pathological signaling pathways, human induced pluripotent stem cell (hiPSCs)-derived motor neurons (MNs) from FUS- and SOD1 ALS patients and healthy controls were systematically compared to independent published datasets. Here through this study we created a gene profile of ALS by analyzing the DEGs, the Kyoto encyclopedia of Genes and Genomes (KEGG) pathways, the interactome and the transcription factor profiles (TF) that would identify altered molecular/functional signatures and their interactions at both transcriptional (mRNAs) and translational levels (hub proteins and TFs). Our findings suggest that FUS and SOD1 may develop from dysregulation in several unique pathways and herpes simplex virus (HSV) infection was among the topmost predominant cellular pathways connected to FUS and not to SOD1. In contrast, SOD1 is mainly characterized by alterations in the metabolic pathways and alterations in the neuroactive-ligand–receptor interactions. This suggests that different genetic ALS forms are singular diseases rather than part of a common spectrum. This is important for patient stratification clearly pointing towards the need for individualized medicine approaches in ALS. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Graphical abstract

16 pages, 1866 KiB  
Article
SCH23390 Reduces Methamphetamine Self-Administration and Prevents Methamphetamine-Induced Striatal LTD
by Yosef Avchalumov, Wulfran Trenet, Juan Piña-Crespo and Chitra Mandyam
Int. J. Mol. Sci. 2020, 21(18), 6491; https://doi.org/10.3390/ijms21186491 - 05 Sep 2020
Cited by 16 | Viewed by 3451
Abstract
Extended-access methamphetamine self-administration results in unregulated intake of the drug; however, the role of dorsal striatal dopamine D1-like receptors (D1Rs) in the reinforcing properties of methamphetamine under extended-access conditions is unclear. Acute (ex vivo) and chronic (in vivo) methamphetamine [...] Read more.
Extended-access methamphetamine self-administration results in unregulated intake of the drug; however, the role of dorsal striatal dopamine D1-like receptors (D1Rs) in the reinforcing properties of methamphetamine under extended-access conditions is unclear. Acute (ex vivo) and chronic (in vivo) methamphetamine exposure induces neuroplastic changes in the dorsal striatum, a critical region implicated in instrumental learning. For example, methamphetamine exposure alters high-frequency stimulation (HFS)-induced long-term depression in the dorsal striatum; however, the effect of methamphetamine on HFS-induced long-term potentiation (LTP) in the dorsal striatum is unknown. In the current study, dorsal striatal infusion of SCH23390, a D1R antagonist, prior to extended-access methamphetamine self-administration reduced methamphetamine addiction-like behavior. Reduced behavior was associated with reduced expression of PSD-95 in the dorsal striatum. Electrophysiological findings demonstrate that superfusion of methamphetamine reduced basal synaptic transmission and HFS-induced LTP in dorsal striatal slices, and SCH23390 prevented this effect. These results suggest that alterations in synaptic transmission and synaptic plasticity induced by acute methamphetamine via D1Rs could assist with methamphetamine-induced modification of corticostriatal circuits underlying the learning of goal-directed instrumental actions and formation of habits, mediating escalation of methamphetamine self-administration and methamphetamine addiction-like behavior. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

11 pages, 1951 KiB  
Article
Sephin1 Protects Neurons against Excitotoxicity Independently of the Integrated Stress Response
by Asier Ruiz, Jone Zuazo, Carolina Ortiz-Sanz, Celia Luchena, Carlos Matute and Elena Alberdi
Int. J. Mol. Sci. 2020, 21(17), 6088; https://doi.org/10.3390/ijms21176088 - 24 Aug 2020
Cited by 8 | Viewed by 2542
Abstract
Sephin1 is a derivative of guanabenz that inhibits the dephosphorylation of the eukaryotic initiation factor 2 alpha (eIF2α) and therefore may enhance the integrated stress response (ISR), an adaptive mechanism against different cellular stresses, such as accumulation of misfolded proteins. Unlike guanabenz, Sephin1 [...] Read more.
Sephin1 is a derivative of guanabenz that inhibits the dephosphorylation of the eukaryotic initiation factor 2 alpha (eIF2α) and therefore may enhance the integrated stress response (ISR), an adaptive mechanism against different cellular stresses, such as accumulation of misfolded proteins. Unlike guanabenz, Sephin1 provides neuroprotection without adverse effects on the α2-adrenergic system and therefore it is considered a promising pharmacological therapeutic tool. Here, we have studied the effects of Sephin1 on N-methyl-D-aspartic acid (NMDA) receptor signaling which may modulate the ISR and contribute to excitotoxic neuronal loss in several neurodegenerative conditions. Time-course analysis of peIF2α levels after NMDA receptor overactivation showed a delayed dephosphorylation that occurred in the absence of activating transcription factor 4 (ATF4) and therefore independently of the ISR, in contrast to that observed during endoplasmic reticulum (ER) stress induced by tunicamycin and thapsigargin. Similar to guanabenz, Sephin1 completely blocked NMDA-induced neuronal death and was ineffective against AMPA-induced excitotoxicity, whereas it did not protect from experimental ER stress. Interestingly, both guanabenz and Sephin1 partially but significantly reduced NMDA-induced cytosolic Ca2+ increase, leading to a complete inhibition of subsequent calpain activation. We conclude that Sephin1 and guanabenz share common strong anti-excitotoxic properties with therapeutic potential unrelated to the ISR. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

22 pages, 4980 KiB  
Article
Vascular Remodeling in Moyamoya Angiopathy: From Peripheral Blood Mononuclear Cells to Endothelial Cells
by Francesca Tinelli, Sara Nava, Francesco Arioli, Gloria Bedini, Emma Scelzo, Daniela Lisini, Giuseppe Faragò, Andrea Gioppo, Elisa F. Ciceri, Francesco Acerbi, Paolo Ferroli, Ignazio G. Vetrano, Silvia Esposito, Veronica Saletti, Chiara Pantaleoni, Federica Zibordi, Nardo Nardocci, Maria Luisa Zedde, Alessandro Pezzini, Vincenzo Di Lazzaro, Fioravante Capone, Maria Luisa Dell’Acqua, Peter Vajkoczy, Elisabeth Tournier-Lasserve, Eugenio A. Parati, Anna Bersano and Laura Gattiadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2020, 21(16), 5763; https://doi.org/10.3390/ijms21165763 - 11 Aug 2020
Cited by 15 | Viewed by 3064
Abstract
The pathophysiological mechanisms of Moyamoya angiopathy (MA), which is a rare cerebrovascular condition characterized by recurrent ischemic/hemorrhagic strokes, are still largely unknown. An imbalance of vasculogenic/angiogenic mechanisms has been proposed as one possible disease aspect. Circulating endothelial progenitor cells (cEPCs) have been hypothesized [...] Read more.
The pathophysiological mechanisms of Moyamoya angiopathy (MA), which is a rare cerebrovascular condition characterized by recurrent ischemic/hemorrhagic strokes, are still largely unknown. An imbalance of vasculogenic/angiogenic mechanisms has been proposed as one possible disease aspect. Circulating endothelial progenitor cells (cEPCs) have been hypothesized to contribute to vascular remodeling of MA, but it remains unclear whether they might be considered a disease effect or have a role in disease pathogenesis. The aim of the present study was to provide a morphological, phenotypical, and functional characterization of the cEPCs from MA patients to uncover their role in the disease pathophysiology. cEPCs were identified from whole blood as CD45dimCD34+CD133+ mononuclear cells. Morphological, biochemical, and functional assays were performed to characterize cEPCs. A significant reduced level of cEPCs was found in blood samples collected from a homogeneous group of adult (mean age 46.86 ± 11.7; 86.36% females), Caucasian, non-operated MA patients with respect to healthy donors (HD; p = 0.032). Since no difference in cEPC characteristics and functionality was observed between MA patients and HD, a defective recruitment mechanism could be involved in the disease pathophysiology. Collectively, our results suggest that cEPC level more than endothelial progenitor cell (EPC) functionality seems to be a potential marker of MA. The validation of our results on a larger population and the correlation with clinical data as well as the use of more complex cellular model could help our understanding of EPC role in MA pathophysiology. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

18 pages, 2879 KiB  
Article
DNA Methyltransferase 1 (DNMT1) Acts on Neurodegeneration by Modulating Proteostasis-Relevant Intracellular Processes
by Cathrin Bayer, Georg Pitschelatow, Nina Hannemann, Jenice Linde, Julia Reichard, Daniel Pensold and Geraldine Zimmer-Bensch
Int. J. Mol. Sci. 2020, 21(15), 5420; https://doi.org/10.3390/ijms21155420 - 30 Jul 2020
Cited by 16 | Viewed by 3624
Abstract
The limited regenerative capacity of neurons requires a tightly orchestrated cell death and survival regulation in the context of longevity, as well as age-associated and neurodegenerative diseases. Subordinate to genetic networks, epigenetic mechanisms, such as DNA methylation and histone modifications, are involved in [...] Read more.
The limited regenerative capacity of neurons requires a tightly orchestrated cell death and survival regulation in the context of longevity, as well as age-associated and neurodegenerative diseases. Subordinate to genetic networks, epigenetic mechanisms, such as DNA methylation and histone modifications, are involved in the regulation of neuronal functionality and emerge as key contributors to the pathophysiology of neurodegenerative diseases. DNA methylation, a dynamic and reversible process, is executed by DNA methyltransferases (DNMTs). DNMT1 was previously shown to act on neuronal survival in the aged brain, whereby a DNMT1-dependent modulation of processes relevant for protein degradation was proposed as an underlying mechanism. Properly operating proteostasis networks are a mandatory prerequisite for the functionality and long-term survival of neurons. Malfunctioning proteostasis is found, inter alia, in neurodegenerative contexts. Here, we investigated whether DNMT1 affects critical aspects of the proteostasis network by a combination of expression studies, live cell imaging, and protein biochemical analyses. We found that DNMT1 negatively impacts retrograde trafficking and autophagy, with both being involved in the clearance of aggregation-prone proteins by the aggresome–autophagy pathway. In line with this, we found that the transport of GFP-labeled mutant huntingtin (HTT) to perinuclear regions, proposed to be cytoprotective, also depends on DNMT1. Depletion of Dnmt1 accelerated perinuclear HTT aggregation and improved the survival of cells transfected with mutant HTT. This suggests that mutant HTT-induced cytotoxicity is at least in part mediated by DNMT1-dependent modulation of degradative pathways. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

18 pages, 4383 KiB  
Article
ORAI2 Down-Regulation Potentiates SOCE and Decreases Aβ42 Accumulation in Human Neuroglioma Cells
by Elena Scremin, Mario Agostini, Alessandro Leparulo, Tullio Pozzan, Elisa Greotti and Cristina Fasolato
Int. J. Mol. Sci. 2020, 21(15), 5288; https://doi.org/10.3390/ijms21155288 - 25 Jul 2020
Cited by 14 | Viewed by 3123
Abstract
Senile plaques, the hallmarks of Alzheimer’s Disease (AD), are generated by the deposition of amyloid-beta (Aβ), the proteolytic product of amyloid precursor protein (APP), by β and γ-secretase. A large body of evidence points towards a role for Ca2+ imbalances in the [...] Read more.
Senile plaques, the hallmarks of Alzheimer’s Disease (AD), are generated by the deposition of amyloid-beta (Aβ), the proteolytic product of amyloid precursor protein (APP), by β and γ-secretase. A large body of evidence points towards a role for Ca2+ imbalances in the pathophysiology of both sporadic and familial forms of AD (FAD). A reduction in store-operated Ca2+ entry (SOCE) is shared by numerous FAD-linked mutations, and SOCE is involved in Aβ accumulation in different model cells. In neurons, both the role and components of SOCE remain quite obscure, whereas in astrocytes, SOCE controls their Ca2+-based excitability and communication to neurons. Glial cells are also directly involved in Aβ production and clearance. Here, we focus on the role of ORAI2, a key SOCE component, in modulating SOCE in the human neuroglioma cell line H4. We show that ORAI2 overexpression reduces both SOCE level and stores Ca2+ content, while ORAI2 downregulation significantly increases SOCE amplitude without affecting store Ca2+ handling. In Aβ-secreting H4-APPswe cells, SOCE inhibition by BTP2 and SOCE augmentation by ORAI2 downregulation respectively increases and decreases Aβ42 accumulation. Based on these findings, we suggest ORAI2 downregulation as a potential tool to rescue defective SOCE in AD, while preventing plaque formation. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Graphical abstract

Review

Jump to: Editorial, Research

38 pages, 824 KiB  
Review
Signalling Pathways Implicated in Alzheimer′s Disease Neurodegeneration in Individuals with and without Down Syndrome
by Carmen Martínez-Cué and Noemí Rueda
Int. J. Mol. Sci. 2020, 21(18), 6906; https://doi.org/10.3390/ijms21186906 - 20 Sep 2020
Cited by 16 | Viewed by 3961
Abstract
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology [...] Read more.
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer’s disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

23 pages, 3043 KiB  
Review
Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases
by Alazne Arrazola Sastre, Miriam Luque Montoro, Patricia Gálvez-Martín, Hadriano M Lacerda, Alejandro Lucia, Francisco Llavero and José Luis Zugaza
Int. J. Mol. Sci. 2020, 21(17), 6312; https://doi.org/10.3390/ijms21176312 - 31 Aug 2020
Cited by 44 | Viewed by 6878
Abstract
Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating [...] Read more.
Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs). Moreover, small GTPases transduce signals by their downstream effector molecules. Many studies demonstrate that small GTPases of the Ras family are involved in neurodegeneration processes. Here, in this review, we focus on the signaling pathways controlled by these small protein superfamilies that culminate in neurodegenerative pathologies, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Specifically, we concentrate on the two most studied families of the Ras superfamily: the Ras and Rho families. We summarize the latest findings of small GTPases of the Ras and Rho families in neurodegeneration in order to highlight these small proteins as potential therapeutic targets capable of slowing down different neurodegenerative diseases. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

25 pages, 1952 KiB  
Review
Bile Acid Signaling in Neurodegenerative and Neurological Disorders
by Stephanie M. Grant and Sharon DeMorrow
Int. J. Mol. Sci. 2020, 21(17), 5982; https://doi.org/10.3390/ijms21175982 - 20 Aug 2020
Cited by 82 | Viewed by 10363
Abstract
Bile acids are commonly known as digestive agents for lipids. The mechanisms of bile acids in the gastrointestinal track during normal physiological conditions as well as hepatic and cholestatic diseases have been well studied. Bile acids additionally serve as ligands for signaling molecules [...] Read more.
Bile acids are commonly known as digestive agents for lipids. The mechanisms of bile acids in the gastrointestinal track during normal physiological conditions as well as hepatic and cholestatic diseases have been well studied. Bile acids additionally serve as ligands for signaling molecules such as nuclear receptor Farnesoid X receptor and membrane-bound receptors, Takeda G-protein-coupled bile acid receptor and sphingosine-1-phosphate receptor 2. Recent studies have shown that bile acid signaling may also have a prevalent role in the central nervous system. Some bile acids, such as tauroursodeoxycholic acid and ursodeoxycholic acid, have shown neuroprotective potential in experimental animal models and clinical studies of many neurological conditions. Alterations in bile acid metabolism have been discovered as potential biomarkers for prognosis tools as well as the expression of various bile acid receptors in multiple neurological ailments. This review explores the findings of recent studies highlighting bile acid-mediated therapies and bile acid-mediated signaling and the roles they play in neurodegenerative and neurological diseases. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

18 pages, 2320 KiB  
Review
R-Ras GTPases Signaling Role in Myelin Neurodegenerative Diseases
by Berta Alcover-Sanchez, Gonzalo Garcia-Martin, Francisco Wandosell and Beatriz Cubelos
Int. J. Mol. Sci. 2020, 21(16), 5911; https://doi.org/10.3390/ijms21165911 - 17 Aug 2020
Cited by 16 | Viewed by 4924
Abstract
Myelination is required for fast and efficient synaptic transmission in vertebrates. In the central nervous system, oligodendrocytes are responsible for creating myelin sheaths that isolate and protect axons, even throughout adulthood. However, when myelin is lost, the failure of remyelination mechanisms can cause [...] Read more.
Myelination is required for fast and efficient synaptic transmission in vertebrates. In the central nervous system, oligodendrocytes are responsible for creating myelin sheaths that isolate and protect axons, even throughout adulthood. However, when myelin is lost, the failure of remyelination mechanisms can cause neurodegenerative myelin-associated pathologies. From oligodendrocyte progenitor cells to mature myelinating oligodendrocytes, myelination is a highly complex process that involves many elements of cellular signaling, yet many of the mechanisms that coordinate it, remain unknown. In this review, we will focus on the three major pathways involved in myelination (PI3K/Akt/mTOR, ERK1/2-MAPK, and Wnt/β-catenin) and recent advances describing the crosstalk elements which help to regulate them. In addition, we will review the tight relation between Ras GTPases and myelination processes and discuss its potential as novel elements of crosstalk between the pathways. A better understanding of the crosstalk elements orchestrating myelination mechanisms is essential to identify new potential targets to mitigate neurodegeneration. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

17 pages, 2101 KiB  
Review
Reactive Glia Inflammatory Signaling Pathways and Epilepsy
by Pascual Sanz and Maria Adelaida Garcia-Gimeno
Int. J. Mol. Sci. 2020, 21(11), 4096; https://doi.org/10.3390/ijms21114096 - 08 Jun 2020
Cited by 90 | Viewed by 6452
Abstract
Neuroinflammation and epilepsy are interconnected. Brain inflammation promotes neuronal hyper-excitability and seizures, and dysregulation in the glia immune-inflammatory function is a common factor that predisposes or contributes to the generation of seizures. At the same time, acute seizures upregulate the production of pro-inflammatory [...] Read more.
Neuroinflammation and epilepsy are interconnected. Brain inflammation promotes neuronal hyper-excitability and seizures, and dysregulation in the glia immune-inflammatory function is a common factor that predisposes or contributes to the generation of seizures. At the same time, acute seizures upregulate the production of pro-inflammatory cytokines in microglia and astrocytes, triggering a downstream cascade of inflammatory mediators. Therefore, epileptic seizures and inflammatory mediators form a vicious positive feedback loop, reinforcing each other. In this work, we have reviewed the main glial signaling pathways involved in neuroinflammation, how they are affected in epileptic conditions, and the therapeutic opportunities they offer to prevent these disorders. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

12 pages, 639 KiB  
Review
S100A6 and Its Brain Ligands in Neurodegenerative Disorders
by Anna Filipek and Wiesława Leśniak
Int. J. Mol. Sci. 2020, 21(11), 3979; https://doi.org/10.3390/ijms21113979 - 01 Jun 2020
Cited by 18 | Viewed by 2836
Abstract
The S100A6 protein is present in different mammalian cells and tissues including the brain. It binds Ca2+ and Zn2+ and interacts with many target proteins/ligands. The best characterized ligands of S100A6, expressed at high level in the brain, include CacyBP/SIP and [...] Read more.
The S100A6 protein is present in different mammalian cells and tissues including the brain. It binds Ca2+ and Zn2+ and interacts with many target proteins/ligands. The best characterized ligands of S100A6, expressed at high level in the brain, include CacyBP/SIP and Sgt1. Research concerning the functional role of S100A6 and these two ligands indicates that they are involved in various signaling pathways that regulate cell proliferation, differentiation, cytoskeletal organization, and others. In this review, we focused on the expression/localization of these proteins in the brain and on their possible role in neurodegenerative diseases. Published results demonstrate that S100A6, CacyBP/SIP, and Sgt1 are expressed in various brain structures and in the spinal cord and can be found in different cell types including neurons and astrocytes. When it comes to their possible involvement in nervous system pathology, it is evident that their expression/level and/or subcellular localization is changed when compared to normal conditions. Among diseases in which such changes have been observed are Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), epileptogenesis, Parkinson’s disease (PD), Huntington’s disease (HD), and others. Full article
(This article belongs to the Special Issue Cell Signaling in Neurodegeneration)
Show Figures

Figure 1

Back to TopTop