ijms-logo

Journal Browser

Journal Browser

Neurodegenerative Disease: From Molecular Basis to Therapy

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 2023) | Viewed by 44115

Printed Edition Available!
A printed edition of this Special Issue is available here.

Special Issue Editor


E-Mail Website
Guest Editor
Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
Interests: molecular biology; neurogenetics; motor neuron diseases; amyotrophic lateral sclerosis; gene variants; gene regulation
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues, 

Neurodegenerative diseases are a heterogeneous group of disorders, largely age-dependent, affecting the central nervous system and eventually leading to neurodegeneration. The prevalence of these diseases is increasing, partly due to the aging population, with a consequent growing economic burden on healthcare systems. The current treatments are mostly symptomatic, without affecting the underlying cause of disease, and have no or only slight effects on disease progression.

Recent advancements in neurobiology and neurogenetics have provided valuable insights into the pathogenesis of neurodegenerative diseases. This has opened the way for the development of molecularly targeted therapies, able to pause or slow the fundamental pathological processes that cause neuronal damage and consequent cognitive and motor dysfunctions. In some cases, neurodegenerative diseases are caused by genetic variants and/or cellular pathway dysregulation. Some mechanisms common to several neurodegenerative conditions have been identified, such as the presence of misfolded protein aggregates, abnormal accumulation of proteins, RNA toxicity, or translational products from repeats expansion within genes. As advances are made in understanding critical aspects of the underlying molecular pathophysiology, therapeutic strategies continue to evolve. Among these, gene therapy is arousing great interest, due to the possibility to deliver functional genetic material to cells, to correct a defective gene.

This Special Issue aims to provide an updated overview of the advancements in research on neurodegenerative diseases, from the understanding of the molecular bases to the development of new therapies. Contributions related, but not limited to Alzheimer's disease and other dementias, Parkinson's disease and Motor Neuron diseases are welcomed, including original research articles and full and mini-reviews.

Dr. Claudia Ricci
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

  • neurodegenerative diseases
  • molecular biology
  • neurogenetics
  • pathogenesis
  • therapy
  • personalized medicine

Related Special Issue

Published Papers (20 papers)

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

Editorial

Jump to: Research, Review, Other

7 pages, 205 KiB  
Editorial
Neurodegenerative Disease: From Molecular Basis to Therapy
by Claudia Ricci
Int. J. Mol. Sci. 2024, 25(2), 967; https://doi.org/10.3390/ijms25020967 - 12 Jan 2024
Viewed by 1659
Abstract
Neurodegenerative diseases are a heterogeneous group of age-related disorders characterised by the progressive degeneration or death of neurons in the central or peripheral nervous system [...] Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)

Research

Jump to: Editorial, Review, Other

17 pages, 3013 KiB  
Article
Simufilam Reverses Aberrant Receptor Interactions of Filamin A in Alzheimer’s Disease
by Hoau-Yan Wang, Erika Cecon, Julie Dam, Zhe Pei, Ralf Jockers and Lindsay H. Burns
Int. J. Mol. Sci. 2023, 24(18), 13927; https://doi.org/10.3390/ijms241813927 - 11 Sep 2023
Cited by 2 | Viewed by 4463
Abstract
Simufilam is a novel oral drug candidate in Phase 3 clinical trials for Alzheimer’s disease (AD) dementia. This small molecule binds an altered form of filamin A (FLNA) that occurs in AD. This drug action disrupts FLNA’s aberrant linkage to the α7 nicotinic [...] Read more.
Simufilam is a novel oral drug candidate in Phase 3 clinical trials for Alzheimer’s disease (AD) dementia. This small molecule binds an altered form of filamin A (FLNA) that occurs in AD. This drug action disrupts FLNA’s aberrant linkage to the α7 nicotinic acetylcholine receptor (α7nAChR), thereby blocking soluble amyloid beta1–42 (Aβ42)’s signaling via α7nAChR that hyperphosphorylates tau. Here, we aimed to clarify simufilam’s mechanism. We now show that simufilam reduced Aβ42 binding to α7nAChR with a 10-picomolar IC50 using time-resolved fluorescence resonance energy transfer (TR-FRET), a robust technology to detect highly sensitive molecular interactions. We also show that FLNA links to multiple inflammatory receptors in addition to Toll-like receptor 4 (TLR4) in postmortem human AD brains and in AD transgenic mice: TLR2, C-X-C chemokine receptor type 4 (CXCR4), C-C chemokine receptor type 5 (CCR5), and T-cell co-receptor cluster of differentiation 4 (CD4). These aberrant FLNA linkages, which can be induced in a healthy control brain by Aβ42 incubation, were disrupted by simufilam. Simufilam reduced inflammatory cytokine release from Aβ42-stimulated human astrocytes. In the AD transgenic mice, CCR5–G protein coupling was elevated, indicating persistent activation. Oral simufilam reduced both the FLNA–CCR5 linkage and the CCR5–G protein coupling in these mice, while restoring CCR5′s responsivity to C-C chemokine ligand 3 (CCL3). By disrupting aberrant FLNA–receptor interactions critical to AD pathogenic pathways, simufilam may promote brain health. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

15 pages, 1736 KiB  
Article
Repeated Methamphetamine Administration Results in Axon Loss Prior to Somatic Loss of Substantia Nigra Pars Compacta and Locus Coeruleus Neurons in Male but Not Female Mice
by Alexander Pilski and Steven M. Graves
Int. J. Mol. Sci. 2023, 24(17), 13039; https://doi.org/10.3390/ijms241713039 - 22 Aug 2023
Cited by 1 | Viewed by 891
Abstract
Methamphetamine (meth) is a neurotoxic psychostimulant that increases monoamine oxidase (MAO)-dependent mitochondrial oxidant stress in axonal but not somatic compartments of substantia nigra pars compacta (SNc) and locus coeruleus (LC) neurons. Chronic meth administration results in the degeneration of SNc and LC neurons [...] Read more.
Methamphetamine (meth) is a neurotoxic psychostimulant that increases monoamine oxidase (MAO)-dependent mitochondrial oxidant stress in axonal but not somatic compartments of substantia nigra pars compacta (SNc) and locus coeruleus (LC) neurons. Chronic meth administration results in the degeneration of SNc and LC neurons in male mice, and MAO inhibition is neuroprotective, suggesting that the deleterious effects of chronic meth begin in axons before advancing to the soma of SNc and LC neurons. To test this hypothesis, mice were administered meth (5 mg/kg) for 14, 21, or 28 days, and SNc and LC axonal lengths and numbers of neurons were quantified. In male mice, the SNc and LC axon lengths decreased with 14, 21, and 28 days of meth, whereas somatic loss was only observed after 28 days of meth; MAO inhibition (phenelzine; 20 mg/kg) prevented axonal and somatic loss of SNc and LC neurons. In contrast, chronic (28-day) meth had no effect on the axon length or numbers of SNc or LC neurons in female mice. The results demonstrate that repeated exposure to meth produces SNc and LC axonal deficits prior to somatic loss in male subjects, consistent with a dying-back pattern of degeneration, whereas female mice are resistant to chronic meth-induced degeneration. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

14 pages, 8912 KiB  
Article
The ε-Isozyme of Protein Kinase C (PKCε) Is Impaired in ALS Motor Cortex and Its Pulse Activation by Bryostatin-1 Produces Long Term Survival in Degenerating SOD1-G93A Motor Neuron-like Cells
by Valentina La Cognata, Agata Grazia D’Amico, Grazia Maugeri, Giovanna Morello, Maria Guarnaccia, Benedetta Magrì, Eleonora Aronica, Daniel L. Alkon, Velia D’Agata and Sebastiano Cavallaro
Int. J. Mol. Sci. 2023, 24(16), 12825; https://doi.org/10.3390/ijms241612825 - 15 Aug 2023
Cited by 1 | Viewed by 1311
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and ultimately fatal neurodegenerative disease, characterized by a progressive depletion of upper and lower motor neurons (MNs) in the brain and spinal cord. The aberrant regulation of several PKC-mediated signal transduction pathways in ALS has [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and ultimately fatal neurodegenerative disease, characterized by a progressive depletion of upper and lower motor neurons (MNs) in the brain and spinal cord. The aberrant regulation of several PKC-mediated signal transduction pathways in ALS has been characterized so far, describing either impaired expression or altered activity of single PKC isozymes (α, β, ζ and δ). Here, we detailed the distribution and cellular localization of the ε-isozyme of protein kinase C (PKCε) in human postmortem motor cortex specimens and reported a significant decrease in both PKCε mRNA (PRKCE) and protein immunoreactivity in a subset of sporadic ALS patients. We furthermore investigated the steady-state levels of both pan and phosphorylated PKCε in doxycycline-activated NSC-34 cell lines carrying the human wild-type (WT) or mutant G93A SOD1 and the biological long-term effect of its transient agonism by Bryostatin-1. The G93A-SOD1 cells showed a significant reduction of the phosphoPKCε/panPKCε ratio compared to the WT. Moreover, a brief pulse activation of PKCε by Bryostatin-1 produced long-term survival in activated G93A-SOD1 degenerating cells in two different cell death paradigms (serum starvation and chemokines-induced toxicity). Altogether, the data support the implication of PKCε in ALS pathophysiology and suggests its pharmacological modulation as a potential neuroprotective strategy, at least in a subgroup of sporadic ALS patients. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

16 pages, 3002 KiB  
Article
Molecular Changes in the Brain of the Wintering Calidris pusilla in the Mangroves of the Amazon River Estuary
by Patrick Douglas Corrêa Pereira, Ediely Pereira Henrique, Emanuel Ramos da Costa, Anderson de Jesus Falcão, Mauro André Damasceno de Melo, Maria Paula Cruz Schneider, Rommel Mario Rodriguez Burbano, Daniel Guerreiro Diniz, Nara Gyzely de Morais Magalhães, David Francis Sherry, Cristovam Wanderley Picanço Diniz and Cristovam Guerreiro-Diniz
Int. J. Mol. Sci. 2023, 24(16), 12712; https://doi.org/10.3390/ijms241612712 - 12 Aug 2023
Viewed by 1967
Abstract
Migrant birds prepare differently to fly north for breeding in the spring and for the flight to lower latitudes during autumn, avoiding the cold and food shortages of the Northern Hemisphere’s harsh winter. The molecular events associated with these fundamental stages in the [...] Read more.
Migrant birds prepare differently to fly north for breeding in the spring and for the flight to lower latitudes during autumn, avoiding the cold and food shortages of the Northern Hemisphere’s harsh winter. The molecular events associated with these fundamental stages in the life history of migrants include the differential gene expression in different tissues. Semipalmated sandpipers (Calidris pusilla) are Arctic-breeding shorebirds that migrate to the coast of South America during the non-breeding season. In a previous study, we demonstrated that between the beginning and the end of the wintering period, substantial glial changes and neurogenesis occur in the brain of C. pusilla. These changes follow the epic journey of the autumn migration when a 5-day non-stop transatlantic flight towards the coast of South America and the subsequent preparation for the long-distance flight of the spring migration takes place. Here, we tested the hypothesis that the differential gene expressions observed in the brains of individuals captured in the autumn and spring windows are consistent with the previously described cellular changes. We searched for differential gene expressions in the brain of the semipalmated sandpiper, of recently arrived birds (RA) from the autumnal migration, and that of individuals in the premigratory period (PM) in the spring. All individuals were collected in the tropical coastal of northern Brazil in the mangrove region of the Amazon River estuary. We generated a de novo neurotranscriptome for C. pusilla individuals and compared the gene expressions across libraries. To that end, we mapped an RNA-Seq that reads to the C. pusilla neurotranscriptome in four brain samples of each group and found that the differential gene expressions in newly arrived and premigratory birds were related with neurogenesis, metabolic pathways (ketone body biosynthetic and the catabolic and lipid biosynthetic processes), and glial changes (astrocyte-dopaminergic neuron signaling, astrocyte differentiation, astrocyte cell migration, and astrocyte activation involved in immune response), as well as genes related to the immune response to virus infections (Type I Interferons), inflammatory cytokines (IL-6, IL-1β, TNF, and NF-κB), NLRP3 inflammasome, anti-inflammatory cytokines (IL-10), and cell death pathways (pyroptosis- and caspase-related changes). Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

13 pages, 7516 KiB  
Article
α5-GABAA Receptor Modulation Reverses Behavioral and Neurophysiological Correlates of Psychosis in Rats with Ventral Hippocampal Alzheimer’s Disease-like Pathology
by Nicole E. Eassa, Stephanie M. Perez, Angela M. Boley, Hannah B. Elam, Dishary Sharmin, James M. Cook and Daniel J. Lodge
Int. J. Mol. Sci. 2023, 24(14), 11788; https://doi.org/10.3390/ijms241411788 - 22 Jul 2023
Cited by 1 | Viewed by 1390
Abstract
Of the 35 million people in the world suffering from Alzheimer’s Disease (AD), up to half experience comorbid psychosis. Antipsychotics, used to treat psychosis, are contraindicated in elderly patients because they increase the risk of premature death. Reports indicate that the hippocampus is [...] Read more.
Of the 35 million people in the world suffering from Alzheimer’s Disease (AD), up to half experience comorbid psychosis. Antipsychotics, used to treat psychosis, are contraindicated in elderly patients because they increase the risk of premature death. Reports indicate that the hippocampus is hyperactive in patients with psychosis and those with AD. Preclinical studies have demonstrated that the ventral hippocampus (vHipp) can regulate dopamine system function, which is thought to underlie symptoms of psychosis. A viral-mediated approach was used to express mutated human genes known to contribute to AD pathology: the Swedish (K670N, M671L), Florida (I716V), and London (V717I) mutations of amyloid precursor protein and two mutations (M146L and L286V) of presenilin 1 specifically in the vHipp, to investigate the selective contribution of AD-like pathology in this region. We observed a significant increase in dopamine neuron population activity and behavioral deficits in this AD-AAV model that mimics observations in rodent models with psychosis-like symptomatologies. Further, systemic administration of MP-III-022 (α5-GABAA receptor selective positive allosteric modulator) was able to reverse aberrant dopamine system function in AD-AAV rats. This study provides evidence for the development of drugs that target α5-GABAA receptors for patients with AD and comorbid psychosis. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

17 pages, 1483 KiB  
Article
The QPLEX™ Plus Assay Kit for the Early Clinical Diagnosis of Alzheimer’s Disease
by Hunjong Na, Ki Young Shin, Dokyung Lee, Changsik Yoon, Sun-Ho Han, Jong-Chan Park, Inhee Mook-Jung, Jisung Jang and Sunghoon Kwon
Int. J. Mol. Sci. 2023, 24(13), 11119; https://doi.org/10.3390/ijms241311119 - 5 Jul 2023
Cited by 1 | Viewed by 1240
Abstract
We recently developed a multiplex diagnostic kit, QPLEX™ Alz plus assay kit, which captures amyloid-β1-40, galectin-3 binding protein, angiotensin-converting enzyme, and periostin simultaneously using microliters of peripheral blood and utilizes an optimized algorithm for screening Alzheimer’s disease (AD) by correlating with cerebral amyloid [...] Read more.
We recently developed a multiplex diagnostic kit, QPLEX™ Alz plus assay kit, which captures amyloid-β1-40, galectin-3 binding protein, angiotensin-converting enzyme, and periostin simultaneously using microliters of peripheral blood and utilizes an optimized algorithm for screening Alzheimer’s disease (AD) by correlating with cerebral amyloid deposition. Owing to the demand for early AD detection, we investigate the potential of our kit for the early clinical diagnosis of AD. A total of 1395 participants were recruited, and their blood samples were analyzed with the QPLEX™ kit. The average of QPLEX™ algorithm values in each group increased gradually in the order of the clinical progression continuum of AD: cognitively normal (0.382 ± 0.150), subjective cognitive decline (0.452 ± 0.130), mild cognitive impairment (0.484 ± 0.129), and AD (0.513 ± 0.136). The algorithm values between each group showed statistically significant differences among groups divided by Mini-Mental State Examination and Clinical Dementia Rating. The QPLEX™ algorithm values could be used to distinguish the clinical continuum of AD or cognitive function. Because blood-based diagnosis is more accessible, convenient, and cost- and time-effective than cerebral spinal fluid or positron emission tomography imaging-based diagnosis, the QPLEX™ kit can potentially be used for health checkups and the early clinical diagnosis of AD. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

16 pages, 3834 KiB  
Article
LAR Downregulation Protects the Astrocytic U251 and Cocultured SH-SY5Y Cells in a Rotenone-Induced Parkinson’s Disease Cell Model
by Wei Zheng, Xiao Han, Bing Han, Gang Li, Jing Gan, Tian Wang, Bo Xu, Jie He, Wenxiao Du, Xiaolin Cao and Zhenhua Wang
Int. J. Mol. Sci. 2023, 24(13), 11111; https://doi.org/10.3390/ijms241311111 - 5 Jul 2023
Cited by 2 | Viewed by 1365
Abstract
Leukocyte common antigen-related protein tyrosine phosphatase (LAR) is a member of the protein tyrosine phosphatase family that serves as a key regulator of cellular survival. It is also involved in neurodevelopment and brain disorders. This study was designed to investigate the role of [...] Read more.
Leukocyte common antigen-related protein tyrosine phosphatase (LAR) is a member of the protein tyrosine phosphatase family that serves as a key regulator of cellular survival. It is also involved in neurodevelopment and brain disorders. This study was designed to investigate the role of LAR in a cell-based model of Parkinson’s disease (PD) in which U251 and SH-SY5Y cells were used as models of astrocytes and dopaminergic neurons, respectively. Cell viability, cell death, cell morphology, protein phosphorylation and expression, ATP levels, reactive oxygen species (ROS) generation, and mitochondrial membrane potential were analyzed in the wild-type (WT) and heterozygous LAR-knockout astrocytoma U251 cells to assess the cell state, signal transduction, and mitochondrial function. LAR downregulation showed a protective effect in rotenone-exposed U251 cells by increasing cell viability, reducing cell mortality, and restoring appropriate cellular morphology. LAR downregulation enhanced IGF-1R phosphorylation and downstream signal transduction as evidenced by increases in the Akt and GSK-3β phosphorylation, as well as the upregulation of NRF2 and HO-1. The downregulation of LAR also augmented DJ-1 levels in these cells. The enhanced Akt and GSK-3β phosphorylation contributed to a reduced Bax/Bcl2 ratio and suppressed apoptosis after rotenone exposure. Heterozygous LAR-knockout U251 cells exhibited higher mitochondrial function evidenced by increased mitochondrial membrane potential, ATP contents, and reduced ROS production compared to the WT cells following rotenone exposure. Further studies showed that the astrocytic protection mediated by the heterozygous knockout of LAR was associated with the activation of Akt. A specific Akt inhibitor, MK2206, reduced the cell viability, Akt and GSK3β phosphorylation, and HO-1 and NRF2 expression in U251 cells exposed to rotenone. Astrocytes provide structural and metabolic support to maintain neuronal health. Astrocytic glial cell-derived neurotrophic factor (GDNF) production is vital for dopaminergic neuron survival. Heterozygous LAR-knockout U251 cells produced higher amounts of GDNF than the WT cells. The SH-SY5Y cells cocultured with heterozygous LAR-knockout U251 cells exhibited greater viability than that of cells cocultured with WT U251 cells in response to rotenone. Together, these findings demonstrate that the heterozygous knockout of LAR in astrocytes can play a key role in protecting both astrocytic cells and cocultured neurons in a rotenone-induced cell-based model of PD. This neuroprotective effect is attributable to the augmentation of IGF1R-Akt-GDNF signaling and the maintenance of astrocytic mitochondrial function. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

17 pages, 4923 KiB  
Article
ARL6IP5 Ameliorates α-Synuclein Burden by Inducing Autophagy via Preventing Ubiquitination and Degradation of ATG12
by Ibrar Siddique, Kajal Kamble, Sakshi Gupta, Kavita Solanki, Sumnil Bhola, Nuzhat Ahsan and Sarika Gupta
Int. J. Mol. Sci. 2023, 24(13), 10499; https://doi.org/10.3390/ijms241310499 - 22 Jun 2023
Cited by 3 | Viewed by 1346
Abstract
Recent advanced studies in neurodegenerative diseases have revealed several links connecting autophagy and neurodegeneration. Autophagy is the major cellular degradation process for the removal of toxic protein aggregates responsible for neurodegenerative diseases. More than 30 autophagy-related proteins have been identified as directly participating [...] Read more.
Recent advanced studies in neurodegenerative diseases have revealed several links connecting autophagy and neurodegeneration. Autophagy is the major cellular degradation process for the removal of toxic protein aggregates responsible for neurodegenerative diseases. More than 30 autophagy-related proteins have been identified as directly participating in the autophagy process. Proteins regulating the process of autophagy are much more numerous and unknown. To address this, in our present study, we identified a novel regulator (ARL6IP5) of neuronal autophagy and showed that the level of ARL6IP5 decreases in the brain with age and in Parkinson’s disease in mice and humans. Moreover, a cellular model of PD (Wild type and A53T mutant α-synuclein overexpression) has also shown decreased levels of ARL6IP5. ARL6IP5 overexpression reduces α-synuclein aggregate burden and improves cell survival in an A53T model of Parkinson’s disease. Interestingly, detailed mechanistic studies revealed that ARL6IP5 is an autophagy inducer. ARL6IP5 enhances Rab1-dependent autophagosome initiation and elongation by stabilizing free ATG12. We report for the first time that α-synuclein downregulates ARL6IP5 to inhibit autophagy-dependent clearance of toxic aggregates that exacerbate neurodegeneration. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

16 pages, 2718 KiB  
Article
Neuroprotection of Andrographolide against Neurotoxin MPP+-Induced Apoptosis in SH-SY5Y Cells via Activating Mitophagy, Autophagy, and Antioxidant Activities
by Prachayaporn Prasertsuksri, Pichnaree Kraokaew, Kanta Pranweerapaiboon, Prasert Sobhon and Kulathida Chaithirayanon
Int. J. Mol. Sci. 2023, 24(10), 8528; https://doi.org/10.3390/ijms24108528 - 10 May 2023
Cited by 2 | Viewed by 2445
Abstract
Parkinson’s disease (PD) is associated with dopaminergic neuron loss and alpha-synuclein aggregation caused by ROS overproduction, leading to mitochondrial dysfunction and autophagy impairment. Recently, andrographolide (Andro) has been extensively studied for various pharmacological properties, such as anti-diabetic, anti-cancer, anti-inflammatory, and anti-atherosclerosis. However, its [...] Read more.
Parkinson’s disease (PD) is associated with dopaminergic neuron loss and alpha-synuclein aggregation caused by ROS overproduction, leading to mitochondrial dysfunction and autophagy impairment. Recently, andrographolide (Andro) has been extensively studied for various pharmacological properties, such as anti-diabetic, anti-cancer, anti-inflammatory, and anti-atherosclerosis. However, its potential neuroprotective effects on neurotoxin MPP+-induced SH-SY5Y cells, a cellular PD model, remain uninvestigated. In this study, we hypothesized that Andro has neuroprotective effects against MPP+-induced apoptosis, which may be mediated through the clearance of dysfunctional mitochondria by mitophagy and ROS by antioxidant activities. Herein, Andro pretreatment could attenuate MPP+-induced neuronal cell death that was reflected by reducing mitochondrial membrane potential (MMP) depolarization, alpha-synuclein, and pro-apoptotic proteins expressions. Concomitantly, Andro attenuated MPP+-induced oxidative stress through mitophagy, as indicated by increasing colocalization of MitoTracker Red with LC3, upregulations of the PINK1–Parkin pathway, and autophagy-related proteins. On the contrary, Andro-activated autophagy was compromised when pretreated with 3-MA. Furthermore, Andro activated the Nrf2/KEAP1 pathway, leading to increasing genes encoding antioxidant enzymes and activities. This study elucidated that Andro exhibited significant neuroprotective effects against MPP+-induced SH-SY5Y cell death in vitro by enhancing mitophagy and clearance of alpha-synuclein through autophagy, as well as increasing antioxidant capacity. Our results provide evidence that Andro could be considered a potential supplement for PD prevention. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

13 pages, 1617 KiB  
Article
NAADP-Evoked Ca2+ Signaling Leads to Mutant Huntingtin Aggregation and Autophagy Impairment in Murine Astrocytes
by Cássia Arruda de Souza Pereira, Natalia de Castro Medaglia, Rodrigo Portes Ureshino, Claudia Bincoletto, Manuela Antonioli, Gian Maria Fimia, Mauro Piacentini, Gustavo José da Silva Pereira, Adolfo Garcia Erustes and Soraya Soubhi Smaili
Int. J. Mol. Sci. 2023, 24(6), 5593; https://doi.org/10.3390/ijms24065593 - 15 Mar 2023
Cited by 5 | Viewed by 1526
Abstract
Huntington’s disease (HD) is a progressive neurodegenerative disease characterized by mutations in the huntingtin gene (mHtt), causing an unstable repeat of the CAG trinucleotide, leading to abnormal long repeats of polyglutamine (poly-Q) in the N-terminal region of the huntingtin, which form abnormal conformations [...] Read more.
Huntington’s disease (HD) is a progressive neurodegenerative disease characterized by mutations in the huntingtin gene (mHtt), causing an unstable repeat of the CAG trinucleotide, leading to abnormal long repeats of polyglutamine (poly-Q) in the N-terminal region of the huntingtin, which form abnormal conformations and aggregates. Alterations in Ca2+ signaling are involved in HD models and the accumulation of mutated huntingtin interferes with Ca2+ homeostasis. Lysosomes are intracellular Ca2+ storages that participate in endocytic and lysosomal degradation processes, including autophagy. Nicotinic acid adenine dinucleotide phosphate (NAADP) is an intracellular second messenger that promotes Ca2+ release from the endo-lysosomal system via Two-Pore Channels (TPCs) activation. Herein, we show the impact of lysosomal Ca2+ signals on mHtt aggregation and autophagy blockade in murine astrocytes overexpressing mHtt-Q74. We observed that mHtt-Q74 overexpression causes an increase in NAADP-evoked Ca2+ signals and mHtt aggregation, which was inhibited in the presence of Ned-19, a TPC antagonist, or BAPTA-AM, a Ca2+ chelator. Additionally, TPC2 silencing revert the mHtt aggregation. Furthermore, mHtt has been shown co-localized with TPC2 which may contribute to its effects on lysosomal homeostasis. Moreover, NAADP-mediated autophagy was also blocked since its function is dependent on lysosomal functionality. Taken together, our data show that increased levels of cytosolic Ca2+ mediated by NAADP causes mHtt aggregation. Additionally, mHtt co-localizes with the lysosomes, where it possibly affects organelle functions and impairs autophagy. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Graphical abstract

25 pages, 2336 KiB  
Article
Adaptive Stimulations in a Biophysical Network Model of Parkinson’s Disease
by Thomas Stojsavljevic, Yixin Guo and Dominick Macaluso
Int. J. Mol. Sci. 2023, 24(6), 5555; https://doi.org/10.3390/ijms24065555 - 14 Mar 2023
Cited by 1 | Viewed by 1408
Abstract
Deep brain stimulation (DBS)—through a surgically implanted electrode to the subthalamic nucleus (STN)—has become a widely used therapeutic option for the treatment of Parkinson’s disease and other neurological disorders. The standard conventional high-frequency stimulation (HF) that is currently used has several drawbacks. To [...] Read more.
Deep brain stimulation (DBS)—through a surgically implanted electrode to the subthalamic nucleus (STN)—has become a widely used therapeutic option for the treatment of Parkinson’s disease and other neurological disorders. The standard conventional high-frequency stimulation (HF) that is currently used has several drawbacks. To overcome the limitations of HF, researchers have been developing closed-loop and demand-controlled, adaptive stimulation protocols wherein the amount of current that is delivered is turned on and off in real-time in accordance with a biophysical signal. Computational modeling of DBS in neural network models is an increasingly important tool in the development of new protocols that aid researchers in animal and clinical studies. In this computational study, we seek to implement a novel technique of DBS where we stimulate the STN in an adaptive fashion using the interspike time of the neurons to control stimulation. Our results show that our protocol eliminates bursts in the synchronized bursting neuronal activity of the STN, which is hypothesized to cause the failure of thalamocortical neurons (TC) to respond properly to excitatory cortical inputs. Further, we are able to significantly decrease the TC relay errors, representing potential therapeutics for Parkinson’s disease. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

16 pages, 3757 KiB  
Article
Origin of Elevated S-Glutathionylated GAPDH in Chronic Neurodegenerative Diseases
by Paul A. Hyslop, Leonard N. Boggs and Michael O. Chaney
Int. J. Mol. Sci. 2023, 24(6), 5529; https://doi.org/10.3390/ijms24065529 - 14 Mar 2023
Cited by 3 | Viewed by 1312
Abstract
H2O2-oxidized glyceraldehyde-3-phosphate dehydrogenase (GAPDH) catalytic cysteine residues (Cc(SH) undergo rapid S-glutathionylation. Restoration of the enzyme activity is accomplished by thiol/disulfide SN2 displacement (directly or enzymatically) forming glutathione disulfide (G(SS)G) and active enzyme, a process that [...] Read more.
H2O2-oxidized glyceraldehyde-3-phosphate dehydrogenase (GAPDH) catalytic cysteine residues (Cc(SH) undergo rapid S-glutathionylation. Restoration of the enzyme activity is accomplished by thiol/disulfide SN2 displacement (directly or enzymatically) forming glutathione disulfide (G(SS)G) and active enzyme, a process that should be facile as Cc(SH) reside on the subunit surface. As S-glutathionylated GAPDH accumulates following ischemic and/or oxidative stress, in vitro/silico approaches have been employed to address this paradox. Cc(SH) residues were selectively oxidized and S-glutathionylated. Kinetics of GAPDH dehydrogenase recovery demonstrated that glutathione is an ineffective reactivator of S-glutathionylated GAPDH compared to dithiothreitol. Molecular dynamic simulations (MDS) demonstrated strong binding interactions between local residues and S-glutathione. A second glutathione was accommodated for thiol/disulfide exchange forming a tightly bound glutathione disulfide G(SS)G. The proximal sulfur centers of G(SS)G and Cc(SH) remained within covalent bonding distance for thiol/disulfide exchange resonance. Both these factors predict inhibition of dissociation of G(SS)G, which was verified by biochemical analysis. MDS also revealed that both S-glutathionylation and bound G(SS)G significantly perturbed subunit secondary structure particularly within the S-loop, region which interacts with other cellular proteins and mediates NAD(P)+ binding specificity. Our data provides a molecular rationale for how oxidative stress elevates S-glutathionylated GAPDH in neurodegenerative diseases and implicates novel targets for therapeutic intervention. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

17 pages, 1150 KiB  
Review
Amyloid Precursor Protein and Alzheimer’s Disease
by Kseniia S. Orobets and Andrey L. Karamyshev
Int. J. Mol. Sci. 2023, 24(19), 14794; https://doi.org/10.3390/ijms241914794 - 30 Sep 2023
Cited by 3 | Viewed by 2459
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative disorders associated with age or inherited mutations. It is characterized by severe dementia in the late stages that affect memory, cognitive functions, and daily life overall. AD progression is linked to the accumulation [...] Read more.
Alzheimer’s disease (AD) is one of the most common neurodegenerative disorders associated with age or inherited mutations. It is characterized by severe dementia in the late stages that affect memory, cognitive functions, and daily life overall. AD progression is linked to the accumulation of cytotoxic amyloid beta (Aβ) and hyperphosphorylated tau protein combined with other pathological features such as synaptic loss, defective energy metabolism, imbalances in protein, and metal homeostasis. Several treatment options for AD are under investigation, including antibody-based therapy and stem cell transplantation. Amyloid precursor protein (APP) is a membrane protein considered to play a main role in AD pathology. It is known that APP in physiological conditions follows a non-amyloidogenic pathway; however, it can proceed to an amyloidogenic scenario, which leads to the generation of extracellular deleterious Aβ plaques. Not all steps of APP biogenesis are clear so far, and these questions should be addressed in future studies. AD is a complex chronic disease with many factors that contribute to disease progression. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

16 pages, 1312 KiB  
Review
Vitamin E and Its Molecular Effects in Experimental Models of Neurodegenerative Diseases
by Bianca Caroline da Cunha Germano, Lara Cristina Carlos de Morais, Francisca Idalina Neta, Amélia Carolina Lopes Fernandes, Francisco Irochima Pinheiro, Amália Cinthia Meneses do Rego, Irami Araújo Filho, Eduardo Pereira de Azevedo, José Rodolfo Lopes de Paiva Cavalcanti, Fausto Pierdona Guzen and Ricardo Ney Cobucci
Int. J. Mol. Sci. 2023, 24(13), 11191; https://doi.org/10.3390/ijms241311191 - 7 Jul 2023
Cited by 7 | Viewed by 2203
Abstract
With the advancement of in vivo studies and clinical trials, the pathogenesis of neurodegenerative diseases has been better understood. However, gaps still need to be better elucidated, which justifies the publication of reviews that explore the mechanisms related to the development of these [...] Read more.
With the advancement of in vivo studies and clinical trials, the pathogenesis of neurodegenerative diseases has been better understood. However, gaps still need to be better elucidated, which justifies the publication of reviews that explore the mechanisms related to the development of these diseases. Studies show that vitamin E supplementation can protect neurons from the damage caused by oxidative stress, with a positive impact on the prevention and progression of neurodegenerative diseases. Thus, this review aims to summarize the scientific evidence of the effects of vitamin E supplementation on neuroprotection and on neurodegeneration markers in experimental models. A search for studies published between 2000 and 2023 was carried out in the PubMed, Web of Science, Virtual Health Library (BVS), and Embase databases, in which the effects of vitamin E in experimental models of neurodegeneration were investigated. A total of 5669 potentially eligible studies were identified. After excluding the duplicates, 5373 remained, of which 5253 were excluded after checking the titles, 90 articles after reading the abstracts, and 11 after fully reviewing the manuscripts, leaving 19 publications to be included in this review. Experiments with in vivo models of neurodegenerative diseases demonstrated that vitamin E supplementation significantly improved memory, cognition, learning, motor function, and brain markers associated with neuroregeneration and neuroprotection. Vitamin E supplementation reduced beta-amyloid (Aβ) deposition and toxicity in experimental models of Alzheimer’s disease. In addition, it decreased tau-protein hyperphosphorylation and increased superoxide dismutase and brain-derived neurotrophic factor (BDNF) levels in rodents, which seems to indicate the potential use of vitamin E in preventing and delaying the progress of degenerative lesions in the central nervous system. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Graphical abstract

14 pages, 1263 KiB  
Review
Cellular Protein Aggregates: Formation, Biological Effects, and Ways of Elimination
by Jun-Hao Wen, Xiang-Hong He, Ze-Sen Feng, Dong-Yi Li, Ji-Xin Tang and Hua-Feng Liu
Int. J. Mol. Sci. 2023, 24(10), 8593; https://doi.org/10.3390/ijms24108593 - 11 May 2023
Cited by 8 | Viewed by 3672
Abstract
The accumulation of protein aggregates is the hallmark of many neurodegenerative diseases. The dysregulation of protein homeostasis (or proteostasis) caused by acute proteotoxic stresses or chronic expression of mutant proteins can lead to protein aggregation. Protein aggregates can interfere with a variety of [...] Read more.
The accumulation of protein aggregates is the hallmark of many neurodegenerative diseases. The dysregulation of protein homeostasis (or proteostasis) caused by acute proteotoxic stresses or chronic expression of mutant proteins can lead to protein aggregation. Protein aggregates can interfere with a variety of cellular biological processes and consume factors essential for maintaining proteostasis, leading to a further imbalance of proteostasis and further accumulation of protein aggregates, creating a vicious cycle that ultimately leads to aging and the progression of age-related neurodegenerative diseases. Over the long course of evolution, eukaryotic cells have evolved a variety of mechanisms to rescue or eliminate aggregated proteins. Here, we will briefly review the composition and causes of protein aggregation in mammalian cells, systematically summarize the role of protein aggregates in the organisms, and further highlight some of the clearance mechanisms of protein aggregates. Finally, we will discuss potential therapeutic strategies that target protein aggregates in the treatment of aging and age-related neurodegenerative diseases. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

20 pages, 1249 KiB  
Review
Appropriate Macronutrients or Mineral Elements Are Beneficial to Improve Depression and Reduce the Risk of Depression
by Zhengyang Quan, Hui Li, Zhenzhen Quan and Hong Qing
Int. J. Mol. Sci. 2023, 24(8), 7098; https://doi.org/10.3390/ijms24087098 - 12 Apr 2023
Cited by 5 | Viewed by 3135
Abstract
Depression is a common mental disorder that seriously affects the quality of life and leads to an increasing global suicide rate. Macro, micro, and trace elements are the main components that maintain normal physiological functions of the brain. Depression is manifested in abnormal [...] Read more.
Depression is a common mental disorder that seriously affects the quality of life and leads to an increasing global suicide rate. Macro, micro, and trace elements are the main components that maintain normal physiological functions of the brain. Depression is manifested in abnormal brain functions, which are considered to be tightly related to the imbalance of elements. Elements associated with depression include glucose, fatty acids, amino acids, and mineral elements such as lithium, zinc, magnesium, copper, iron, and selenium. To explore the relationship between these elements and depression, the main literature in the last decade was mainly searched and summarized on PubMed, Google Scholar, Scopus, Web of Science, and other electronic databases with the keywords “depression, sugar, fat, protein, lithium, zinc, magnesium, copper, iron, and selenium”. These elements aggravate or alleviate depression by regulating a series of physiological processes, including the transmission of neural signals, inflammation, oxidative stress, neurogenesis, and synaptic plasticity, which thus affect the expression or activity of physiological components such as neurotransmitters, neurotrophic factors, receptors, cytokines, and ion-binding proteins in the body. For example, excessive fat intake can lead to depression, with possible mechanisms including inflammation, increased oxidative stress, reduced synaptic plasticity, and decreased expression of 5-Hydroxytryptamine (5-HT), Brain Derived Neurotrophic Factor (BDNF), Postsynaptic density protein 95(PSD-95), etc. Supplementing mineral elements, such as selenium, zinc, magnesium, or lithium as a psychotropic medication is mostly used as an auxiliary method to improve depression with other antidepressants. In general, appropriate nutritional elements are essential to treat depression and prevent the risk of depression. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

23 pages, 1009 KiB  
Review
New Pathways Identify Novel Drug Targets for the Prevention and Treatment of Alzheimer’s Disease
by Botond Penke, Mária Szűcs and Ferenc Bogár
Int. J. Mol. Sci. 2023, 24(6), 5383; https://doi.org/10.3390/ijms24065383 - 11 Mar 2023
Cited by 8 | Viewed by 5053
Abstract
Alzheimer’s disease (AD) is an incurable, progressive neurodegenerative disorder. AD is a complex and multifactorial disease that is responsible for 60–80% of dementia cases. Aging, genetic factors, and epigenetic changes are the main risk factors for AD. Two aggregation-prone proteins play a decisive [...] Read more.
Alzheimer’s disease (AD) is an incurable, progressive neurodegenerative disorder. AD is a complex and multifactorial disease that is responsible for 60–80% of dementia cases. Aging, genetic factors, and epigenetic changes are the main risk factors for AD. Two aggregation-prone proteins play a decisive role in AD pathogenesis: β-amyloid (Aβ) and hyperphosphorylated tau (pTau). Both of them form deposits and diffusible toxic aggregates in the brain. These proteins are the biomarkers of AD. Different hypotheses have tried to explain AD pathogenesis and served as platforms for AD drug research. Experiments demonstrated that both Aβ and pTau might start neurodegenerative processes and are necessary for cognitive decline. The two pathologies act in synergy. Inhibition of the formation of toxic Aβ and pTau aggregates has been an old drug target. Recently, successful Aβ clearance by monoclonal antibodies has raised new hopes for AD treatments if the disease is detected at early stages. More recently, novel targets, e.g., improvements in amyloid clearance from the brain, application of small heat shock proteins (Hsps), modulation of chronic neuroinflammation by different receptor ligands, modulation of microglial phagocytosis, and increase in myelination have been revealed in AD research. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

Other

67 pages, 6516 KiB  
Perspective
The Amyloid Cascade Hypothesis 2.0 for Alzheimer’s Disease and Aging-Associated Cognitive Decline: From Molecular Basis to Effective Therapy
by Vladimir Volloch and Sophia Rits-Volloch
Int. J. Mol. Sci. 2023, 24(15), 12246; https://doi.org/10.3390/ijms241512246 - 31 Jul 2023
Cited by 9 | Viewed by 1181
Abstract
With the long-standing amyloid cascade hypothesis (ACH) largely discredited, there is an acute need for a new all-encompassing interpretation of Alzheimer’s disease (AD). Whereas such a recently proposed theory of AD is designated ACH2.0, its commonality with the ACH is limited to the [...] Read more.
With the long-standing amyloid cascade hypothesis (ACH) largely discredited, there is an acute need for a new all-encompassing interpretation of Alzheimer’s disease (AD). Whereas such a recently proposed theory of AD is designated ACH2.0, its commonality with the ACH is limited to the recognition of the centrality of amyloid-β (Aβ) in the disease, necessitated by the observation that all AD-causing mutations affect, in one way or another, Aβ. Yet, even this narrow commonality is superficial since AD-causing Aβ of the ACH differs distinctly from that specified in the ACH2.0: Whereas in the former, the disease is caused by secreted extracellular Aβ, in the latter, it is triggered by Aβ-protein-precursor (AβPP)-derived intraneuronal Aβ (iAβ) and driven by iAβ generated independently of AβPP. The ACH2.0 envisions AD as a two-stage disorder. The first, asymptomatic stage is a decades-long accumulation of AβPP-derived iAβ, which occurs via internalization of secreted Aβ and through intracellular retention of a fraction of Aβ produced by AβPP proteolysis. When AβPP-derived iAβ reaches critical levels, it activates a self-perpetuating AβPP-independent production of iAβ that drives the second, devastating AD stage, a cascade that includes tau pathology and culminates in neuronal loss. The present study analyzes the dynamics of iAβ accumulation in health and disease and concludes that it is the prime factor driving both AD and aging-associated cognitive decline (AACD). It discusses mechanisms potentially involved in AβPP-independent generation of iAβ, provides mechanistic interpretations for all principal aspects of AD and AACD including the protective effect of the Icelandic AβPP mutation, the early onset of FAD and the sequential manifestation of AD pathology in defined regions of the affected brain, and explains why current mouse AD models are neither adequate nor suitable. It posits that while drugs affecting the accumulation of AβPP-derived iAβ can be effective only protectively for AD, the targeted degradation of iAβ is the best therapeutic strategy for both prevention and effective treatment of AD and AACD. It also proposes potential iAβ-degrading drugs. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

10 pages, 1732 KiB  
Concept Paper
Obstetric Neuropathy in Diabetic Patients: The “Double Hit Hypothesis”
by Dieu Thao Nguyen, Mohammad Hooshmand Zaferanieh, Asa C. Black, Jr., Kamron Reza Hamedi, Richard L. Goodwin and Thomas I. Nathaniel
Int. J. Mol. Sci. 2023, 24(7), 6812; https://doi.org/10.3390/ijms24076812 - 6 Apr 2023
Cited by 1 | Viewed by 1605
Abstract
The two-hit model has been proposed to explain the effects of diabetes on mothers who are already in a putative subclinical damaged state and then undergo neuronal damage during the delivery process. However, the anatomical and pathophysiological mechanisms are not well understood. Our [...] Read more.
The two-hit model has been proposed to explain the effects of diabetes on mothers who are already in a putative subclinical damaged state and then undergo neuronal damage during the delivery process. However, the anatomical and pathophysiological mechanisms are not well understood. Our overarching hypothesis in this review paper is that pregnant women who are diabetic have a damaged peripheral nervous system, constituting the “first hit” hypothesis. The delivery process itself—the “second hit”—can produce neurological damage to the mother. Women with diabetes mellitus (DM) are at risk for neurological damage during both hits, but the cumulative effects of both “hits” pose a greater risk of neurological damage and pathophysiological changes during delivery. In our analysis, we introduce the different steps of our concept paper. Subsequently, we describe each of the topics. First, we outline the mechanisms by which diabetes acts as a detrimental variable in neuropathy by focusing on the most common form of diabetic neuropathy, diabetic distal symmetrical polyneuropathy, also known as distal sensorimotor neuropathy. The possible role of macrosomia in causing diabetic neuropathy and obstetric neurological injury is discussed. Second, we describe how vaginal delivery can cause various obstetrical neurological syndromes and pathophysiological changes. Third, we highlight the risk of obstetric neuropathy and discuss anatomical sites at which lesions may occur, including lesions during delivery. Fourth, we characterize the pathophysiological pathways involved in the causation of diabetic neuropathy. Finally, we highlight diabetic damage to sensory vs. motor nerves, including how hyperglycemia causes different types of damage depending on the location of nerve cell bodies. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

Back to TopTop