[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,448)

Search Parameters:
Keywords = amyotrophic lateral sclerosis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 607 KiB  
Review
Understanding Amyotrophic Lateral Sclerosis: Pathophysiology, Diagnosis, and Therapeutic Advances
by Radu Eugen Rizea, Antonio-Daniel Corlatescu, Horia Petre Costin, Adrian Dumitru and Alexandru Vlad Ciurea
Int. J. Mol. Sci. 2024, 25(18), 9966; https://doi.org/10.3390/ijms25189966 (registering DOI) - 15 Sep 2024
Abstract
This review offers an in-depth examination of amyotrophic lateral sclerosis (ALS), addressing its epidemiology, pathophysiology, clinical presentation, diagnostic techniques, and current as well as emerging treatments. The purpose is to condense key findings and illustrate the complexity of ALS, which is shaped by [...] Read more.
This review offers an in-depth examination of amyotrophic lateral sclerosis (ALS), addressing its epidemiology, pathophysiology, clinical presentation, diagnostic techniques, and current as well as emerging treatments. The purpose is to condense key findings and illustrate the complexity of ALS, which is shaped by both genetic and environmental influences. We reviewed the literature to discuss recent advancements in understanding molecular mechanisms such as protein misfolding, mitochondrial dysfunction, oxidative stress, and axonal transport defects, which are critical for identifying potential therapeutic targets. Significant progress has been made in refining diagnostic criteria and identifying biomarkers, leading to earlier and more precise diagnoses. Although current drug treatments provide some benefits, there is a clear need for more effective therapies. Emerging treatments, such as gene therapy and stem cell therapy, show potential in modifying disease progression and improving the quality of life for ALS patients. The review emphasizes the importance of continued research to address challenges such as disease variability and the limited effectiveness of existing treatments. Future research should concentrate on further exploring the molecular foundations of ALS and developing new therapeutic approaches. The implications for clinical practice include ensuring the accessibility of new treatments and that healthcare systems are equipped to support ongoing research and patient care. Full article
(This article belongs to the Special Issue Molecular Research of Amyotrophic Lateral Sclerosis)
Show Figures

Figure 1

Figure 1
<p>Role of Chitinase as a Biomarker in ALS Pathophysiology. This image illustrates how activated microglia and astrocytes in ALS contribute to chitinase expression, which serves as a biomarker for disease progression and therapeutic monitoring.</p>
Full article ">
43 pages, 4610 KiB  
Review
Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases
by Dong Guo, Zhihao Liu, Jinglin Zhou, Chongrong Ke and Daliang Li
Int. J. Mol. Sci. 2024, 25(18), 9947; https://doi.org/10.3390/ijms25189947 (registering DOI) - 15 Sep 2024
Viewed by 186
Abstract
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to [...] Read more.
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to promote neuronal development, establish neural architecture, and shape the central nervous system (CNS), although the role of PCD extends beyond the CNS. Abnormalities in PCD signaling cascades contribute to the irreversible loss of neuronal cells and function, leading to the onset and progression of neurodegenerative diseases. In this review, we summarize the molecular processes and features of different modalities of PCD, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other novel forms of PCD, and their effects on the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Additionally, we examine the key factors involved in these PCD signaling pathways and discuss the potential for their development as therapeutic targets and strategies. Therefore, therapeutic strategies targeting the inhibition or facilitation of PCD signaling pathways offer a promising approach for clinical applications in treating neurodegenerative diseases. Full article
(This article belongs to the Special Issue Cell Apoptosis, 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>The execution mechanisms of apoptosis, necroptosis, and pyroptosis are described in detail. (<b>a</b>) Both the intrinsic and extrinsic pathways of apoptosis are illustrated, highlighting the key signaling molecules and processes involved; (<b>b</b>) the assembly and function of various necroptosome structures are depicted, emphasizing the key proteins and their roles; (<b>c</b>) the pathways of pyroptosis execution are presented, detailing the stimuli and their effects on cellular components. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: AIFs, apoptosis-inducing factors; Apaf-1, apoptosis protease activating factor-1; APP, amyloid precursor protein; ASC, apoptosis-associated speck-like protein containing a CARD; BH3-only proteins, Bcl-2 homology 3 domain only proteins; BID, BH3-interacting-domain death agonist; CARD, caspase recruitment domain; CTCs, circulating tumor cells; Cyt-c, cytochrome-c; DISC, death inducing signaling complex; DR6, death receptor 6; dsDNA, double-stranded DNA; ER, endoplasmic reticulum; FADD, Fas-associated death domain; GSDM, gasdermin; IAPs, inhibitors of apoptosis proteins; IFNARs, interferon alpha receptors; IFNs, interferons; IL-18, interleukin-18; IL-1β, interleukin-1β; K, potassium; LPS, lipopolysaccharide; MLKL, mixed-lineage kinase-like; NK, natural killer; NLR, nucleotide-binding oligomerization domain-like receptor; NLRP3, NLR family pyrin domain containing 3; NLS, nuclear localization signal; PD-1, programmed death 1; PD-L1, programmed cell death-ligand 1; PRRs, pattern recognition receptors; PtpB, protein tyrosine phosphatase B; RHIM, RIP (receptor-interacting protein) homology interaction motifs; RIPK1, serine/threonine protein kinase 1; RIPK3, serine/threonine protein kinase 3; Smac, small mitochondria-derived activator of caspase; SpeB, streptococcal pyrogenic exotoxin B; TAK1, TGF-β-activated kinase 1; T-BID, truncated BID; TLR3, toll-like receptor 3; TLR4, toll-like receptor 4; TRADD, TNF receptor-associated death domain; TRIF, TIR-domain-containing adapter-inducing interferon-β; YopJ, yersinia outer protein J; ZBP1, Z-DNA/RNA-binding protein; Z-dsDNA/RNA, Z-form double-stranded DNA/RNA.</p>
Full article ">Figure 2
<p>The pathways of cuproptosis and ferroptosis are illustrated. (<b>a</b>) Ferroptosis is a form of iron-dependent programmed cell death resulting from intracellular iron overload. The figure depicts the lipid peroxidation induced by dysregulated iron metabolism and the subsequent execution of ferroptosis; (<b>b</b>) abnormal copper metabolism and accumulation can lead to protein toxicity, mitochondrial damage, and cuproptosis. For details, refer to the corresponding section of this article. Abbreviations: Cu, copper; DSF, disulfiram; ES, elesclomol; FDX1, ferredoxin 1; Fe, iron; NCOA4, nuclear receptor coactivator 4; NRAMP2 (also known as SLC11A2), natural resistance-associated macrophage protein 2; ROS, reactive oxygen species; S, sulfur; SLC25A3, solute carrier family 25 member 3; STEAP, six-transmembrane epithelial antigen of prostate; TCA, tricarboxylic acid cycle; TF, transferrin; TFR1, transferrin receptor 1.</p>
Full article ">Figure 3
<p>An overview of the mechanisms of various other forms of PCD. (<b>a</b>) MPT-driven necrosis is mediated by the activation of CYPD and the formation of PTPC, leading to a loss of selective permeability of the inner mitochondrial membrane, resulting in cell swelling and membrane rupture; (<b>b</b>) oxeiptosis is a form of cell death induced by oxygen radicals and mediated by the hyperactivation of the KEAP1-PGAM5-AIFM1 signaling cascade; (<b>c</b>) LDCD is a form of cell death caused by changes in lysosomal membrane permeability, resulting in the leakage of lysosomal contents and subsequent alterations in mitochondrial outer membrane permeability; (<b>d</b>) parthanatos is a form of cell death induced by DNA damage, resulting in the overactivation of PARP1; (<b>e</b>) alkaliptosis is a form of cell death induced by intracellular alkalinization caused by JTC-801, an opioid receptor-like 1 (ORL1) receptor selective antagonist [<a href="#B5-ijms-25-09947" class="html-bibr">5</a>]; (<b>f</b>) in cells with high SLC7A11 expression, increased cystine uptake leads to NADPH depletion, abnormal disulfide bond formation, cytoskeletal collapse, and disulfidptosis; (<b>g</b>) aberrant autophagy leads to excessive ER-phagy, excessive mitophagy, and ADCD. For details, refer to the corresponding section of this article. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: ADCD, autophagy-dependent cell death; AIFM1, apoptosis-inducing factor mitochondria-associated 1; ATP, adenosine triphosphate; ATP6V0D1, ATPase H+ transporting V0 subunit d1; ATPase, adenosine triphosphatase; CA9, carbonic anhydrase 9; CYPD, cyclophilin D; Cyt-c, cytochrome c; DRAM1, DNA damage-regulated autophagy modulator 1; DRP1, dynamin-related protein 1; ER, endoplasmic reticulum; Fe, iron; IMS, intermembrane space; K, potassium; KEAP1, kelch-like ECH-associated protein 1; LDCD, lysosome-dependent cell death; LMP, lysosomal membrane permeabilization; Mito, mitochondria; MOMP, mitochondrial outer membrane permeabilization; MPT, mitochondrial permeability transition; Na, sodium; NADH, nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor κB; PARP1, poly(ADP-ribose) polymerase 1; PGAM, phosphoglycerate mutase; PGAM5, PGAM family member 5; pH, potential of hydrogen; reticulophagy, selective autophagy of the endoplasmic reticulum; PINK1, PTEN-induced kinase 1; PTPC, permeability transition pore complex; ROS, reactive oxygen species; S, sulfur; SLC7A11, solute carrier family 7 member 11; STAT3, signal transducer and activator of transcription 3; UPR, unfolded protein response.</p>
Full article ">Figure 4
<p>The pathways of NETosis and Entosis are depicted. (<b>a</b>) Cellular stress responses induce autophagy, granzyme release and translocation, chromatin decondensation, and cell membrane pore formation, leading to the release of web-like DNA–protein structures and resulting in NETosis; (<b>b</b>) cells undergo entosis, an intracellular cell death process, by inserting themselves into neighboring cells through adhesion proteins. For details, refer to the corresponding section of this article. Abbreviations: Ca, calcium; CTNNA1, catenin alpha 1; ENTosis, entotic cell death; ERK, extracellular signal-regulated kinase; GSDM, gasdermin; GSDMD-N,Gasdermin D N-terminal; LC3, microtubule-associated protein 1 light chain 3; LPS, lipopolysaccharide; MEK, MAP kinase kinase; MPO, myeloperoxidase; mtDNA, mitochondrial DNA; mtROS, mitochondrial reactive oxygen species; NADPH, nicotinamide adenine dinucleotide phosphate; NE, neutrophil elastase; NETosis, neutrophil extracellular trap cell death; NETs, neutrophil extracellular traps; PAD4, peptidylarginine deiminase 4; PKC, protein kinase C; RAF, RAF proto-oncogene serine/threonine-protein kinase; TLR, toll-like receptor.</p>
Full article ">Figure 5
<p>The process of programmed cell death in the development of AD, PD and HD is significant. (<b>a</b>) Various forms of programmed cell death play roles in the pathogenesis and progression of AD, including their effects on Tau protein hyperphosphorylation, Aβ plaque formation, and neuronal cell death; (<b>b</b>) in PD, various forms of PCD induce dopaminergic neuronal loss and death by promoting αSyn aggregation, leading to mitochondrial dysfunction and neuroinflammation. (<b>c</b>) In HD, mHTT induces mitochondrial dysfunction and neuroinflammation by promoting the expression of pro-apoptotic factors and activating necroptosis and ferroptosis. Additionally, the aggregation of mHTT proteins is associated with impaired autophagy, further exacerbating neuronal damage. For details, refer to the corresponding section of this article. The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: AD, Alzheimer’s disease; Akt, protein kinase B; APP, amyloid precursor protein; Aβ, amyloid β; BAX, Bcl-2 associated x-protein; Bcl-2, B-cell lymphoma-2; BDNF, brain-derived neurotrophic factor; BIM, Bcl-2 interacting mediator of cell death; CGA, cytosine-guanine-adenine triplet; CREB, cAMP-response element binding protein; Cu, cuprum; CYPD, cyclophilin D; Cyt-c, cytochrome-c; ER, endoplasmic reticulum; Fe, ferrum; GPX4, glutathione peroxidase 4; GSDMD, gasdermin-D; GSK-3β, glycogen synthase kinase 3β; HD, Huntington’s disease; ICAM-1, intercellular adhesion molecule-1; IL-18, interleukin-18; IL-1β, interleukin-1β; JAK, janus kinase; JNK, c-Jun N-terminal kinase; LFA-1, lymphocyte function-associated antigen 1; LRP1, low-density lipoprotein receptor-related protein 1; LRRK2, leucine-rich repeat kinase 2; MAPK, mitogen-activated protein kinase; mHTT, mutant huntingtin; MLKL, mixed lineage kinase domain-like protein; MOMP, mitochondrial outer membrane permeabilization; MPT, mitochondrial permeability transition; mTOR, mammalian target of rapamycin; NET, neutrophil extracellular traps; NFTs, neurofibrillary tangles; NF-κB, nuclear factor κB; NLRP3, NLR family pyrin domain containing 3; NLR, nucleotide-binding oligomerization domain-like receptor; NO, nitric oxide; PARK7, parkinsonism associated deglycase; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; PD, Parkinson’s disease; PI3K, phosphoinositide 3-kinase; PINK1, PTEN induced kinase 1; polyQ, polyglutamine; PRKN, parkin RBR E3 ubiquitin protein ligase; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; SNCA, alpha-synuclein; αSyn, α-synuclein; STAT, signal transducer and activator of transcription; Tau, microtubule-associated protein Tau; TNF-α, tumor necrosis factor-α; TrkB, tropomyosin receptor kinase B.</p>
Full article ">Figure 6
<p>The role of PCD in the progression of ALS, SMA and MS is critical. The figure illustrates the involvement of PCD pathways in the pathogenesis of ALS, SMA, and MS, highlighting both the factors that contribute to disease progression and those that are beneficial for disease control. Additionally, it describes the common pathways through which PCD exerts its effects across these diseases. For more details, refer to the corresponding section of this article. Abbreviations: ADCD, autophagy-dependent cell death; ALS, amyotrophic lateral sclerosis; Bcl-2, B-cell lymphoma-2; C9ORF72, chromosome 9 open reading frame 72; Cu, cuprum; CYPD, cyclophilin D; Fe, ferrum; FUS, fused in sarcoma/translocated in liposarcoma; GSDMD, gasdermin-D; IL-18, interleukin-18; IL-1β, interleukin-1β; JNK, c-Jun N-terminal kinase; KO, knockout; MPT, mitochondrial permeability transition; mPTP, mitochondrial permeability transition pore; MS, multiple sclerosis; NET, neutrophil extracellular traps; NETosis, neutrophil extracellular trap cell death; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; SMA, spinal muscular atrophy; SMN, survival motor neuron; SOD1, superoxide dismutase 1; TARDBP, TAR DNA-binding protein; TDP-43, TAR DNA-binding protein 43.</p>
Full article ">Figure 7
<p>The role of programmed cell death (PCD) in the progression of traumatic brain injury (TBI) and stroke. The figure illustrates various aspects that contribute to disease progression, revealing the role of PCD in these conditions and considerations beneficial for disease control and treatment. Additionally, it describes the pathways through which PCD exerts its effects both individually and collectively in TBI and stroke. For details, refer to the corresponding section of this article. The upward and downward arrows represent an increase and decrease in content or concentration, respectively. The circular arrow signifies that "apoptosis in neurons of the ischemic penumbra may be recoverable." The red line in the image signifies obstruction or limited functionality. Please refer to the original text for a detailed description of this content. Abbreviations: ADCD, autophagy-dependent cell death; ATP, adenosine triphosphate; Bcl-2, B-cell lymphoma-2; Ca, calcium; CNS, central nervous system; Cu, cuprum; CYPD, cyclophilin D; DRGs, dorsal root ganglions; ER, endoplasmic reticulum; FasL, Fas ligand; Fe, ferrum; HIF-1α, hypoxia-inducible factor 1α; HMGB1, high-mobility group box 1; IL-1β, interleukin-1β; IP, ischemic penumbra; K, kalium; LDCD, lysosome-dependent cell death; Mito, mitochondria; MLKL, mixed lineage kinase domain-like protein; MPT, mitochondrial permeability transition; Na, natrium; NAD, nicotinamide adenine dinucleotide; NET, neutrophil extracellular traps; PAD4, peptidylarginine deiminase 4; PARP1, poly(ADP-ribose) polymerase 1; PCD, programmed cell death; RIPK1, receptor-interacting serine/threonine-protein kinase 1; RIPK3, receptor-interacting serine/threonine-protein kinase 3; ROS, reactive oxygen species; TBI, traumatic brain injury; TNF-α, tumor necrosis factor-α; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand.</p>
Full article ">
18 pages, 404 KiB  
Review
Mycobacterium paratuberculosis: A HERV Turn-On for Autoimmunity, Neurodegeneration, and Cancer?
by Coad Thomas Dow, Ellen S. Pierce and Leonardo A. Sechi
Microorganisms 2024, 12(9), 1890; https://doi.org/10.3390/microorganisms12091890 - 13 Sep 2024
Viewed by 327
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that, over millions of years, became integrated into the human genome. While normally inactive, environmental stimuli such as infections have contributed to the transcriptional reactivation of HERV-promoting pathological conditions, including the development of [...] Read more.
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that, over millions of years, became integrated into the human genome. While normally inactive, environmental stimuli such as infections have contributed to the transcriptional reactivation of HERV-promoting pathological conditions, including the development of autoimmunity, neurodegenerative disease and cancer. What infections trigger HERV activation? Mycobacterium avium subspecies paratuberculosis (MAP) is a pluripotent driver of human disease. Aside from granulomatous diseases, Crohn’s disease, sarcoidosis and Blau syndrome, MAP is associated with autoimmune disease: type one diabetes (T1D), multiple sclerosis (MS), rheumatoid arthritis (RA) and autoimmune thyroiditis. MAP is also associated with Alzheimer’s disease (AD) and Parkinson’s disease (PD). Autoimmune diabetes, MS and RA are the diseases with the strongest MAP/HERV association. There are several other diseases associated with HERV activation, including diseases whose epidemiology and/or pathology would prompt speculation for a causal role of MAP. These include non-solar uveal melanoma, colon cancer, glioblastoma and amyotrophic lateral sclerosis (ALS). This article further points to MAP infection as a contributor to autoimmunity, neurodegenerative disease and cancer via the un-silencing of HERV. We examine the link between the ever-increasing number of MAP-associated diseases and the MAP/HERV intersection with these diverse medical conditions, and propose treatment opportunities based upon this association. Full article
(This article belongs to the Section Medical Microbiology)
26 pages, 1328 KiB  
Review
From Brain to Muscle: The Role of Muscle Tissue in Neurodegenerative Disorders
by Elisa Duranti and Chiara Villa
Biology 2024, 13(9), 719; https://doi.org/10.3390/biology13090719 - 12 Sep 2024
Viewed by 369
Abstract
Neurodegenerative diseases (NDs), like amyotrophic lateral sclerosis (ALS), Alzheimer’s disease (AD), and Parkinson’s disease (PD), primarily affect the central nervous system, leading to progressive neuronal loss and motor and cognitive dysfunction. However, recent studies have revealed that muscle tissue also plays a significant [...] Read more.
Neurodegenerative diseases (NDs), like amyotrophic lateral sclerosis (ALS), Alzheimer’s disease (AD), and Parkinson’s disease (PD), primarily affect the central nervous system, leading to progressive neuronal loss and motor and cognitive dysfunction. However, recent studies have revealed that muscle tissue also plays a significant role in these diseases. ALS is characterized by severe muscle wasting as a result of motor neuron degeneration, as well as alterations in gene expression, protein aggregation, and oxidative stress. Muscle atrophy and mitochondrial dysfunction are also observed in AD, which may exacerbate cognitive decline due to systemic metabolic dysregulation. PD patients exhibit muscle fiber atrophy, altered muscle composition, and α-synuclein aggregation within muscle cells, contributing to motor symptoms and disease progression. Systemic inflammation and impaired protein degradation pathways are common among these disorders, highlighting muscle tissue as a key player in disease progression. Understanding these muscle-related changes offers potential therapeutic avenues, such as targeting mitochondrial function, reducing inflammation, and promoting muscle regeneration with exercise and pharmacological interventions. This review emphasizes the importance of considering an integrative approach to neurodegenerative disease research, considering both central and peripheral pathological mechanisms, in order to develop more effective treatments and improve patient outcomes. Full article
(This article belongs to the Special Issue Repair and Regeneration of Skeletal Muscle)
Show Figures

Figure 1

Figure 1
<p>A schematic representation of skeletal muscle structure. The image was created with the use of Servier Medical Art modified templates, licensed under a Creative Common Attribution 3.0 Unported License (<a href="https://smart.servier.com" target="_blank">https://smart.servier.com</a>, accessed on 28 August 2024).</p>
Full article ">Figure 2
<p>A schematic representation of the accumulation of ROS leading to mitochondrial dysfunction and cellular damage. This cascade of events results in muscle atrophy and weakness. The image was created with the use of Servier Medical Art modified templates, licensed under a Creative Common Attribution 3.0 Unported License (<a href="https://smart.servier.com" target="_blank">https://smart.servier.com</a>, accessed on 28 August 2024).</p>
Full article ">Figure 3
<p>The aggregates of α-synuclein caused by alterations in protein degradation pathways promote the muscle cell alterations typically found in PD patients. The image was created with the use of Servier Medical Art modified templates, licensed under a Creative Common Attribution 3.0 Unported License (<a href="https://smart.servier.com" target="_blank">https://smart.servier.com</a>, accessed on 28 August 2024).</p>
Full article ">
12 pages, 1070 KiB  
Article
Genetic Modifiers of ALS: The Impact of Chromogranin B P413L in a Bulgarian ALS Cohort
by Ivan Tourtourikov, Tihomir Todorov, Teodor Angelov, Teodora Chamova, Ivailo Tournev, Vanyo Mitev and Albena Todorova
Genes 2024, 15(9), 1197; https://doi.org/10.3390/genes15091197 - 12 Sep 2024
Viewed by 321
Abstract
This study investigated the role of the CHGB P413L variant (rs742710) in sporadic amyotrophic lateral sclerosis (sALS) within the Bulgarian population. We analyzed 150 patients with sALS (85 male and 65 female) for the presence of this variant, its potential impact on disease [...] Read more.
This study investigated the role of the CHGB P413L variant (rs742710) in sporadic amyotrophic lateral sclerosis (sALS) within the Bulgarian population. We analyzed 150 patients with sALS (85 male and 65 female) for the presence of this variant, its potential impact on disease susceptibility, and age of onset. Genotyping was performed using PCR amplification and direct Sanger sequencing. Statistical analyses included comparisons with control data from GnomAD v2.1.1, one-way ANOVA, and Kaplan–Meier survival analysis. Results revealed a higher frequency of the minor T allele in patients with sALS compared to all control groups and a statistically significant increase in carrier genotypes compared to non-Finnish Europeans (χ2 = 15.4572, p = 0.000440). However, the impact on age of onset was less clear, with no statistically significant differences observed across genotypes or between carriers and non-carriers of the T allele. Kaplan–Meier analysis suggested a potential 2.5-year-earlier onset in T allele carriers, but the small sample size of carriers limits the reliability of this finding. Our study provides evidence for an association between the CHGB P413L variant and sALS susceptibility in the Bulgarian population, while its effect on age of onset remains uncertain, highlighting the need for further research in larger, diverse cohorts. Full article
(This article belongs to the Section Neurogenomics)
Show Figures

Figure 1

Figure 1
<p>Bar charts depicting the allele frequencies between patients with ALS (blue) and control groups (orange). The minor allele of the rs742710 variant is present with a slightly elevated frequency in the patient group against all tested control populations.</p>
Full article ">Figure 2
<p>Bar charts depicting the allele frequencies between patients with ALS (blue) and control groups (orange). The carrier genotypes C/T and T/T for the rs742710 variant are slightly elevated compared to NFE controls and non-neuro individuals, while only the homozygous T/T genotype has a higher frequency compared to Bulgarian controls and non-neuro individuals.</p>
Full article ">Figure 3
<p>Kaplan–Meier survival curves for onset based on genotype for the entire patient cohort (<b>top</b>) and split by sex (<b>bottom</b>). Heterozygous carriers (orange) show an earlier onset regardless of sex, while homozygous T/T carriers (red) display an earlier onset only in males compared to controls (green).</p>
Full article ">Figure 4
<p>Kaplan–Meier survival curves for onset based on carrier status for the minor allele for the entire patient cohort (<b>top</b>) and split by sex (<b>bottom</b>). Carriers of the T allele (red) present with an earlier onset of the disease, with 2.43 years for the entire cohort, 2.57 years for males and 0.43 years for females compared to controls (green).</p>
Full article ">
18 pages, 3217 KiB  
Article
BV2 Microglial Cell Activation/Polarization Is Influenced by Extracellular Vesicles Released from Mutated SOD1 NSC-34 Motoneuron-like Cells
by Elisabetta Carata, Marco Muci, Stefania Mariano and Elisa Panzarini
Biomedicines 2024, 12(9), 2069; https://doi.org/10.3390/biomedicines12092069 - 11 Sep 2024
Viewed by 311
Abstract
Microglia-mediated neuroinflammation is a key player in the pathogenesis of amyotrophic lateral sclerosis (ALS) as it can contribute to the progressive degeneration of motor neurons (MNs). Here, we investigated the role of mSOD1 NSC-34 MN-like cell-derived extracellular vesicles (EVs) in inducing the activation [...] Read more.
Microglia-mediated neuroinflammation is a key player in the pathogenesis of amyotrophic lateral sclerosis (ALS) as it can contribute to the progressive degeneration of motor neurons (MNs). Here, we investigated the role of mSOD1 NSC-34 MN-like cell-derived extracellular vesicles (EVs) in inducing the activation of BV2 microglial cells. NSC-34-released EVs were isolated by culture medium differential ultracentrifugation to obtain two fractions, one containing small EVs (diameter < 200 nm) and the other containing large EVs (diameter > 200 nm). BV2 cells were incubated with the two EV fractions for 12, 24, and 48 h to evaluate 1) the state of microglial inflammation through RT-PCR of IL-1β, IL-6, IL-4, and IL-10 and 2) the expression of proteins involved in inflammasome activation (IL-β and caspase 1), cell death (caspase 3), and glial cell recruitment (CXCR1), and presence of the TGFβ cytokine receptor (TGFβ-R2). The obtained results suggest a mSOD1 type-dependent polarization of BV2 cells towards an early neurotoxic phenotype and a late neuroprotective status, with an appearance of mixed M1 and M2 microglia subpopulations. A significant role in driving microglial cell activation is played by the TGFβ/CX3CR1 axis. Therefore, targeting the dysregulated microglial response and modulating neuroinflammation could hold promise as a therapeutic strategy for ALS. Full article
(This article belongs to the Special Issue Pharmacological Targets for Neuroinflammation)
Show Figures

Figure 1

Figure 1
<p>Morphological evaluation of BV2 cells cultured for 24 h in the conditioned medium (CM), vesicle-deprived conditioned medium (free EVs CM), small EVs (sEVs), and large EVs (lEVs) of mSOD1 NSC-34 MN-like cells. Left: cell number percentage of dead, activated, and quiescent BV2 cells measured by Nikon Eclipse phase contrast microscope. Right: phase contrast microscope images of BV2 cells. (<b>A</b>) BV2 cells challenged with lEVs from mSOD1G37R NSC-34 cells; (<b>B</b>) quiescent BV2 cells; (<b>C</b>) activated BV2 cells; (<b>D</b>) BV2 cells challenged with sEVs from mSOD1A4V NSC-34 cells; (<b>E</b>) apoptotic BV2 cells. Extracellular vesicles were isolated by differential ultracentrifugation allowing us to obtain two enriched fractions: small EVs (diameter ˂ 200 nm) and large EVs (diameter ˃ 200 nm). Bars: 10 μm. Plasmids used for transfection: pF146 pSOD1WTAcGFP1 (Plasmid #26407); pF147 pSOD1A4VAcGFP1 (Plasmid #26408); pF148 pSOD1G37RAcGFP1 (Plasmid #26409); pF149 pSOD1G85RAcGFP1 (Plasmid #26410); pF150 pSOD1G93AAcGFP1 (Plasmid #26411).</p>
Full article ">Figure 2
<p>TGFβ/CX3CR1 axis regulation. BV2 cells were cultured for 12 (<b>A</b>), 24 (<b>B</b>), and 48 h (<b>C</b>) in the presence of vesicle-deprived conditioned medium (CM-EV), small EVs, and large EVs of mSOD1 NSC-34 MN-like cells. The expression of TGF-β type II receptor (TGFβ-R2), CX3C chemokine receptor 1 (CX3CR1), and interleukin 1β (IL1-β) is normalized to β actin and reported as a relative expression vs. SOD1WT NSC-34 MN-like cells considered as value 1. The values for all experimental groups reported in the histograms represent the means ± SD (n = 3) of three independent experiments; (*) <span class="html-italic">p</span> &lt; 0.05 compared to the control SOD1WT; (**) <span class="html-italic">p</span> &lt; 0.01 compared to the control (wt SOD1). Plasmids used for transfection: pF146 pSOD1WTAcGFP1 (Plasmid #26407); pF147 pSOD1A4VAcGFP1 (Plasmid #26408); pF148 pSOD1G37RAcGFP1 (Plasmid #26409); pF149 pSOD1G85RAcGFP1 (Plasmid #26410); pF150 pSOD1G93AAcGFP1 (Plasmid #26411). Images of representative blots are reported in <a href="#app1-biomedicines-12-02069" class="html-app">Figure S2</a>.</p>
Full article ">Figure 3
<p>Expression of MIF and caspase 1. BV2 cells were cultured for 12 (<b>A</b>), 24 (<b>B</b>), and 48 h (<b>C</b>) in the presence of vesicle-deprived conditioned medium (CM -EV), small EVs, and large EVs of mSOD1 NSC-34 MN-like cells. The expression of MIF and caspase 1 is normalized to β actin and reported as a relative expression vs. SOD1WT NSC-34 MN-like cells considered as value 1. The values for all experimental groups reported in the histograms represent the means ± SD (n = 3) of three independent experiments; (*) <span class="html-italic">p</span> &lt; 0.05 compared to control SOD1WT; (**) <span class="html-italic">p</span> &lt; 0.01 compared to the control (wt SOD1). Plasmids used for transfection: pF146 pSOD1WTAcGFP1 (Plasmid #26407); pF147 pSOD1A4VAcGFP1 (Plasmid #26408); pF148 pSOD1G37RAcGFP1 (Plasmid #26409); pF149 pSOD1G85RAcGFP1 (Plasmid #26410); pF150 pSOD1G93AAcGFP1 (Plasmid #26411). Images of representative blots are reported in <a href="#app1-biomedicines-12-02069" class="html-app">Figure S2</a>.</p>
Full article ">Figure 4
<p>Expression of caspase 3. BV2 cells were cultured for 12 (<b>A</b>), 24 (<b>B</b>), and 48 h (<b>C</b>) in the presence of vesicle-deprived conditioned medium (CM-EV), small EVs, and large EVs of mSOD1 NSC-34 MN-like cells. The expression of caspase 3 is normalized to β actin and reported as a relative expression vs. SOD1WT NSC-34 MN-like cells considered as value 1. The values for all experimental groups reported in the histograms represent the means ± SD (n = 3) of three independent experiments; (*) <span class="html-italic">p</span> &lt; 0.05 compared to control SOD1WT. Plasmids used for transfection: pF146 pSOD1WTAcGFP1 (Plasmid #26407); pF147 pSOD1A4VAcGFP1 (Plasmid #26408); pF148 pSOD1G37RAcGFP1 (Plasmid #26409); pF149 pSOD1G85RAcGFP1 (Plasmid #26410); pF150 pSOD1G93AAcGFP1 (Plasmid #26411). Images of representative blots are reported in <a href="#app1-biomedicines-12-02069" class="html-app">Figure S2</a>.</p>
Full article ">Figure 5
<p>Polarization of BV2 microglial cells. mRNA levels of pro-inflammatory interleukin 1β (IL-1β) and interleukin 6 (IL-6) in BV2 microglial cells challenged for 12 (<b>A</b>), 24 (<b>B</b>), and 48 h (<b>C</b>) with vesicle-deprived conditioned medium (CM-EV), small EVs, and large EVs produced by mSOD1 NSC-34 MN-like cells. Data are normalized to β actin and are expressed as fold levels of mRNA of BV2 cultured with sEVs, lEVs, and free EVs CM vs. mRNA levels of SOD1WTNSC-34 MN-like cells considered as value 1. Values are means ± SD (n = 3); (*) <span class="html-italic">p</span> &lt; 0.05; (**) <span class="html-italic">p</span> &lt; 0.01. Plasmids used for transfection: pF146 pSOD1WTAcGFP1 (Plasmid #26407); pF147 pSOD1A4VAcGFP1 (Plasmid #26408); pF148 pSOD1G37RAcGFP1 (Plasmid #26409); pF149 pSOD1G85RAcGFP1 (Plasmid #26410); pF150 pSOD1G93AAcGFP1 (Plasmid #26411).</p>
Full article ">Figure 6
<p>Polarization of BV2 microglial cells. mRNA levels of pro-inflammatory interleukin 1β (IL-1β) and interleukin 6 (IL-6) in BV2 microglial cells challenged for 12 (<b>A</b>), 24 (<b>B</b>), and 48 h (<b>C</b>) with vesicle-deprived conditioned medium (CM -EV), small EVs, and large EVs produced by mSOD1 NSC-34 MN-like cells. Data are normalized to β actin and are expressed as fold levels of mRNA of BV2 cultured with sEVs, lEVs, and free EVs CM vs. mRNA levels of SOD1WTNSC-34 MN-like cells considered as value 1. Values are means ± SD (n = 3); (*) <span class="html-italic">p</span> &lt; 0.05; (**) <span class="html-italic">p</span> &lt; 0.01. Plasmids used for transfection: pF146 pSOD1WTAcGFP1 (Plasmid #26407); pF147 pSOD1A4VAcGFP1 (Plasmid #26408); pF148 pSOD1G37RAcGFP1 (Plasmid #26409); pF149 pSOD1G85RAcGFP1 (Plasmid #26410); pF150 pSOD1G93AAcGFP1 (Plasmid #26411).</p>
Full article ">
18 pages, 3188 KiB  
Article
Intelligent Control System for Brain-Controlled Mobile Robot Using Self-Learning Neuro-Fuzzy Approach
by Zahid Razzaq, Nihad Brahimi, Hafiz Zia Ur Rehman and Zeashan Hameed Khan
Sensors 2024, 24(18), 5875; https://doi.org/10.3390/s24185875 - 10 Sep 2024
Viewed by 331
Abstract
Brain-computer interface (BCI) provides direct communication and control between the human brain and physical devices. It is achieved by converting EEG signals into control commands. Such interfaces have significantly improved the lives of disabled individuals suffering from neurological disorders—such as stroke, amyotrophic lateral [...] Read more.
Brain-computer interface (BCI) provides direct communication and control between the human brain and physical devices. It is achieved by converting EEG signals into control commands. Such interfaces have significantly improved the lives of disabled individuals suffering from neurological disorders—such as stroke, amyotrophic lateral sclerosis (ALS), and spinal cord injury—by extending their movement range and thereby promoting self-independence. Brain-controlled mobile robots, however, often face challenges in safety and control performance due to the inherent limitations of BCIs. This paper proposes a shared control scheme for brain-controlled mobile robots by utilizing fuzzy logic to enhance safety, control performance, and robustness. The proposed scheme is developed by combining a self-learning neuro-fuzzy (SLNF) controller with an obstacle avoidance controller (OAC). The SLNF controller robustly tracks the user’s intentions, and the OAC ensures the safety of the mobile robot following the BCI commands. Furthermore, SLNF is a model-free controller that can learn as well as update its parameters online, diminishing the effect of disturbances. The experimental results prove the efficacy and robustness of the proposed SLNF controller including a higher task completion rate of 94.29% (compared to 79.29%, and 92.86% for Direct BCI and Fuzzy-PID, respectively), a shorter average task completion time of 85.31 s (compared to 92.01 s and 86.16 s for Direct BCI and Fuzzy-PID, respectively), and reduced settling time and overshoot. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic of the proposed methodology for an EEG controlled mobile robot. The BCI system translates human intentions into steering commands. The shared control system manages automatic switching between user input and autonomous navigation, following <math display="inline"><semantics> <msub> <mi mathvariant="italic">Rule</mi> <mi>X</mi> </msub> </semantics></math> to ensure safety. The robotics system communicates the robot’s states and surrounding information among different controller nodes.</p>
Full article ">Figure 2
<p>Schematic of the wheeled mobile robot showing a two-dimensional coordinate system with the <math display="inline"><semantics> <mrow> <mi>x</mi> <mo>−</mo> <mi>o</mi> <mo>−</mo> <mi>y</mi> </mrow> </semantics></math> global frame and the <math display="inline"><semantics> <mrow> <msub> <mi>x</mi> <mi>c</mi> </msub> <mo>−</mo> <mi>B</mi> <mo>−</mo> <msub> <mi>y</mi> <mi>c</mi> </msub> </mrow> </semantics></math> local frame of the robot. G represents the center of gravity of the robot. The distance between the wheels is <math display="inline"><semantics> <mrow> <mn>2</mn> <mi>R</mi> </mrow> </semantics></math>, and the diameter of each wheel is <math display="inline"><semantics> <mrow> <mn>2</mn> <mi>r</mi> </mrow> </semantics></math>. <math display="inline"><semantics> <mi>ω</mi> </semantics></math> and <span class="html-italic">u</span> represent the angular and linear velocities of the robot, respectively. The angle <math display="inline"><semantics> <mi>ϕ</mi> </semantics></math> indicates the rotation between the global and local frames.</p>
Full article ">Figure 3
<p>Framework of the self−learning neuro−fuzzy control scheme. The reference model filters the desired changes in the plant’s output (w), guiding the plant to follow the set−point trajectory (<span class="html-italic">r</span>). The proportional feedback controller (with <math display="inline"><semantics> <msub> <mi>k</mi> <mi>p</mi> </msub> </semantics></math> gain) minimizes the impact of unmeasured disturbances. The feedback error learning module estimates the correct control signal (<math display="inline"><semantics> <msub> <mover accent="true"> <mi>u</mi> <mo stretchy="false">˜</mo> </mover> <mi>f</mi> </msub> </semantics></math>), while the fuzzy identification scheme updates the controller parameters (ŵ). The feedforward controller (neuro−fuzzy model) approximates the inverse model of a nonlinear plant when properly trained.</p>
Full article ">Figure 4
<p>Structure of Obstacle Avoidance Controller (OAC). IT2FLS processes three LIDAR distance inputs, fuzzifies them into IT2 fuzzy sets, and applies inference rules. The type reducer converts these to IT1 fuzzy sets, and the defuzzifier computes the angular velocity for robot control.</p>
Full article ">Figure 5
<p>The membership functions of AOC for (<b>a</b>) input variables and (<b>b</b>) output variables; Note: LOD = FOD = ROD.</p>
Full article ">Figure 6
<p>Online simulation setup of a robotic system where a user maneuvers the mobile robot using EEG signals to targets A or B, avoiding obstacles.</p>
Full article ">Figure 7
<p>Experimental scenario. The subject focuses on the SSVEP visual stimuli (left screen) to maneuver the robot through obstacles (right screen) and reach the target safely.</p>
Full article ">Figure 8
<p>Comparison of system performance among the Direct BCI, Fuzzy-PID, and SLNF controllers for (<b>a</b>) Task completion rate (%) and (<b>b</b>) Task completion time (seconds). The SLNF controller achieved a higher average task completion rate of 94.29% (vs. 79.29% for Direct BCI and 92.86% for Fuzzy-PID) and a shorter average task completion time of 85.31 s (vs. 92.01 s for Direct BCI and 86.16 s for Fuzzy-PID). Statistically significant differences are indicated with (*) for <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 9
<p>Robot trajectories produced by Subject Three using (<b>a</b>) the proposed SLNF controller and (<b>b</b>) Direct BCI control. The trajectories with the proposed SLNF controller show no collisions, demonstrating the efficacy of OAC, while Direct BCI control method was unable to handle obstacle avoidance.</p>
Full article ">Figure 10
<p>Step input disturbance torque signals. These test the disturbance handling capability of our proposed control system.</p>
Full article ">Figure 11
<p>Disturbance rejection comparison among Direct BCI control, fuzzy-PID, and the proposed controller for (<b>a</b>) linear velocity and (<b>b</b>) angular velocity. Initially, the SLNF controller exhibits the highest overshoot in linear velocity but reduces it over time due to its online learning capability, as shown in the zoomed area. For angular velocity, the SLNF controller shows minimal overshoot and settling time, with disturbance effects decreasing over time. Direct BCI and Fuzzy-PID controllers do not show a significant reduction in disturbance.</p>
Full article ">
13 pages, 3134 KiB  
Article
Evaluation of a Synthetic Retinoid, Ellorarxine, in the NSC-34 Cell Model of Motor Neuron Disease
by Olivia Escudier, Yunxi Zhang, Andrew Whiting and Paul Chazot
Int. J. Mol. Sci. 2024, 25(18), 9764; https://doi.org/10.3390/ijms25189764 - 10 Sep 2024
Viewed by 367
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease worldwide and is characterized by progressive muscle atrophy. There are currently two approved treatments, but they only relieve symptoms briefly and do not cure the disease. The main hindrance to research is [...] Read more.
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease worldwide and is characterized by progressive muscle atrophy. There are currently two approved treatments, but they only relieve symptoms briefly and do not cure the disease. The main hindrance to research is the complex cause of ALS, with its pathogenesis not yet fully elucidated. Retinoids (vitamin A derivatives) appear to be essential in neuronal cells and have been implicated in ALS pathogenesis. This study explores 4-[2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydroquinoxalin-2-yl)ethylnyl]benzoic acid (Ellorarxine, or DC645 or NVG0645), a leading synthetic retinoic acid, discussing its pharmacological mechanisms, neuroprotective properties, and relevance to ALS. The potential therapeutic effect of Ellorarxine was analyzed in vitro using the WT and SOD1G93A NSC-34 cell model of ALS at an administered concentration of 0.3–30 nM. Histological, functional, and biochemical analyses were performed. Elorarxine significantly increased MAP2 expression and neurite length, increased AMPA receptor GluA2 expression and raised intracellular Ca2+ baseline, increased level of excitability, and reduced Ca2+ spike during depolarization in neurites. Ellorarxine also displayed both antioxidant and anti-inflammatory effects. Overall, these results suggest Ellorarxine shows relevance and promise as a novel therapeutic strategy for treatment of ALS. Full article
Show Figures

Figure 1

Figure 1
<p>Serum deprivation reveals the presence of two morphologically distinct populations of WT NSC-34. Phase contrast microscopy images revealed two populations of NSC-34 present in Serum-Free Media (SFM): cells with short neurites (<b>A</b>) and those with longer branching processes (<b>B</b>). Imaging was carried out at one magnification, ×20, on the Zeiss Apotome microscope. Scale bars = 100 μm.</p>
Full article ">Figure 2
<p>(<b>A</b>) Effects of Ellorarxine (DC645) on MAP2 semi-quantitative immunofluorescent staining of differentiated WT NSC-34 cells, (<b>B</b>) Morphological differentiation of undifferentiated NSC-34 cells determined by quantitative measurement of neurite length (α-MEM). (<b>A</b>) Immunofluorescence revealed MAP2 expression was more prevalent in differentiated cells treated with Ellorarxine than without (representative figure from n = 4 replicates). Imaging was carried out at one magnification, ×20, on the Zeiss Apotome microscope. Cells without any primary antibody were used as a negative control. Scale bars = 100 µm. (<b>B</b>) A significant increase is observed for mutant SOD1 NSC-34 cells cultured with Ellorarxine (10 nM). Data expressed as means ± SD. (n = 10) (**** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3
<p>Effects of Ellorarxine (DC645) on CD200 immunofluorescent staining of differentiated WT NSC-34 cells. Staining of both untreated (<b>A</b>), control, cells, and those treated with Ellorarxine (10 nM) (<b>B</b>) for DAPI (left, blue) and CD200 (right, red). Semi-quantitative immunofluorescence revealed CD200 expression was more prevalent in differentiated cells treated with Ellorarxine than without (representative figure from n = 4 replicates). Imaging was carried out at one magnification, ×20, on the Zeiss Apotome microscope. Cells without any primary antibody were used as a negative control. Scale bars = 100 µm.</p>
Full article ">Figure 4
<p>Effects of Ellorarxine (DC645) on TNF-α (<b>A</b>) and IL-6 (<b>B</b>) release of undifferentiated NSC-34 cells incubated with 0.3 µg/mL LPS. NSC-34 cells were pre-treated with 10 nM Ellorarxine for 1 h before 24 h of inflammatory insult with 0.3 µg/mL LPS. Data expressed as means ± SD. (n = 4) (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 5
<p>Effects of Ellorarxine (DC645) on TNF-α release of differentiated NSC-34 cells incubated with LPS. Release of pro-inflammatory signal in differentiated wild type (<b>A</b>) and mutant (<b>B</b>) NSC-34 cells with and without a 1-h pre-incubation with Ellorarxine. Data expressed as means ± SD. (n = 4) (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Effects of Ellorarxine (DC645) on the dose response effect of H<sub>2</sub>O<sub>2</sub> on differentiated wild type and mutant SOD1 NSC-34 cells. Cell viability, compared to control, for cells treated with H<sub>2</sub>O<sub>2</sub> for a 24-h incubation (red) and after a 1-h pre-incubation with Ellorarxine (blue). Arrows indicate the change in IC<sub>50</sub> when treated with Ellorarxine.</p>
Full article ">Figure 7
<p>Effects of Ellorarxine (DC645) on GluA<sub>2</sub> immunofluorescent staining of undifferentiated and differentiated mutant SOD1 NSC-34 cells. Staining of both untreated (<b>A</b>,<b>C</b>), control, cells, and those treated with Ellorarxine (10 nM) (<b>B</b>,<b>D</b>) for DAPI (left, blue) and GluA2 (centre, red) as well as merged channel images (right). Immunofluorescence revealed GluA2 expression was more prevalent in differentiated cells treated with Ellorarxine than not, with clear expression neurite outgrowths (green arrow). Imaging was carried out at one magnification, ×20, on the Zeiss Apotome microscope. Cells without any primary antibody were used as a negative control. Scale bars = 100 µm.</p>
Full article ">Figure 8
<p>Ca<sup>2+</sup> Imaging of differentiated NSC-34 cells: Effects of Ellorarxine (DC645). Changes in normalized fluorescence intensity as a function of time for the average of 10 individual cell body (<b>A</b>), neurite (<b>B</b>). Addition of the depolarizing buffer (KCl) is indicated by an arrow (n = 10).</p>
Full article ">
18 pages, 1569 KiB  
Review
Experimental Cell Models for Investigating Neurodegenerative Diseases
by Cecilia Evangelisti, Sherin Ramadan, Antonio Orlacchio and Emanuele Panza
Int. J. Mol. Sci. 2024, 25(17), 9747; https://doi.org/10.3390/ijms25179747 - 9 Sep 2024
Viewed by 283
Abstract
Experimental models play a pivotal role in biomedical research, facilitating the understanding of disease mechanisms and the development of novel therapeutics. This is particularly true for neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and motor neuron disease, [...] Read more.
Experimental models play a pivotal role in biomedical research, facilitating the understanding of disease mechanisms and the development of novel therapeutics. This is particularly true for neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and motor neuron disease, which present complex challenges for research and therapy development. In this work, we review the recent literature about experimental models and motor neuron disease. We identified three main categories of models that are highly studied by scientists. In fact, experimental models for investigating these diseases encompass a variety of approaches, including modeling the patient’s cell culture, patient-derived induced pluripotent stem cells, and organoids. Each model offers unique advantages and limitations, providing researchers with a range of tools to address complex biological questions. Here, we discuss the characteristics, applications, and recent advancements in terms of each model system, highlighting their contributions to advancing biomedical knowledge and translational research. Full article
(This article belongs to the Special Issue Genetic Bases of Neurodegenerative Disease)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the process to generate experimental models.</p>
Full article ">Figure 2
<p>Primary cell models for investigating neurodegenerative conditions share genes of interest expression profile.</p>
Full article ">
15 pages, 1195 KiB  
Review
Potential Therapeutic Interventions Targeting NAD+ Metabolism for ALS
by Samuel Lundt and Shinghua Ding
Cells 2024, 13(17), 1509; https://doi.org/10.3390/cells13171509 - 9 Sep 2024
Viewed by 577
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting both upper and lower motor neurons. While there have been many potential factors implicated for ALS development, such as oxidative stress and mitochondrial dysfunction, no exact mechanism has been determined at this time. [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting both upper and lower motor neurons. While there have been many potential factors implicated for ALS development, such as oxidative stress and mitochondrial dysfunction, no exact mechanism has been determined at this time. Nicotinamide adenine dinucleotide (NAD+) is one of the most abundant metabolites in mammalian cells and is crucial for a broad range of cellular functions from DNA repair to energy homeostasis. NAD+ can be synthesized from three different intracellular pathways, but it is the NAD+ salvage pathway that generates the largest proportion of NAD+. Impaired NAD+ homeostasis has been connected to aging and neurodegenerative disease-related dysfunctions. In ALS mice, NAD+ homeostasis is potentially disrupted prior to the appearance of physical symptoms and is significantly reduced in the nervous system at the end stage. Treatments targeting NAD+ metabolism, either by administering NAD+ precursor metabolites or small molecules that alter NAD+-dependent enzyme activity, have shown strong beneficial effects in ALS disease models. Here, we review the therapeutic interventions targeting NAD+ metabolism for ALS and their effects on the most prominent pathological aspects of ALS in animal and cell models. Full article
(This article belongs to the Special Issue Genetics and Pathomechanisms of Amyotrophic Lateral Sclerosis (ALS))
Show Figures

Figure 1

Figure 1
<p>NAD<sup>+</sup> salvage pathway and NAD<sup>+</sup>-dependent enzymatic reactions. NAD<sup>+</sup> salvage pathway (<b>top</b>) NAM or NR are converted to NMN by NAMPT or NMRK, respectively. NMNAT generates NAD<sup>+</sup> from NMN. NAD<sup>+</sup> can be reversibly reduced and oxidized or utilized by NADases (SIRTs, PARPs, CD38, and SARM1), which produce NAM as a byproduct that can be re-used to form NAD<sup>+</sup>. NADase reactions (<b>bottom</b>). PARPs add ADPR to substrates. SIRTs remove acetyl groups from target substrates. CD38 and SARM1 generate ADPR/cADPR, which are important for second messenger signaling pathways. All figures were generated using BioRender.com.</p>
Full article ">Figure 2
<p>ALS pathophysiology and the effect of targeted interventions involving NAD<sup>+</sup> metabolism. Treating ALS models with therapeutic interventions altering NAD<sup>+</sup> metabolism ameliorates many disease-related impairments. Mitochondrial dysfunction, oxidative stress response, activation of glial cells, and protein mislocalization, all of which have been hypothesized as being involved in the development of ALS disease, are corrected from these interventions. Additionally, NMJ innervation and function, sites affected early during ALS development, are improved.</p>
Full article ">
16 pages, 1230 KiB  
Review
Expression Changes of miRNAs in Humans and Animal Models of Amyotrophic Lateral Sclerosis and Their Potential Application for Clinical Diagnosis
by Ruili Wang, Liang Chen, Yuning Zhang, Bo Sun and Mengyao Liang
Life 2024, 14(9), 1125; https://doi.org/10.3390/life14091125 - 6 Sep 2024
Viewed by 338
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe motor neuron disease. Current detection methods can only confirm the diagnosis at the onset of the disease, missing the critical window for early treatment. Recent studies using animal models have found that detecting changes in miRNA [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a severe motor neuron disease. Current detection methods can only confirm the diagnosis at the onset of the disease, missing the critical window for early treatment. Recent studies using animal models have found that detecting changes in miRNA sites can predict the onset and severity of the disease in its early stages, facilitating early diagnosis and treatment. miRNAs show expression changes in motor neurons that connect the brain, spinal cord, and brain stem, as well as in the skeletal muscle in mouse models of ALS. Clinically, expression changes in some miRNAs in patients align with those in mouse models, such as the upregulation of miR-29b in the brain and the upregulation of miR-206 in the skeletal muscle. This study provides an overview of some miRNA study findings in humans as well as in animal models, including SOD1, FUS, TDP-43, and C9orf72 transgenic mice and wobbler mice, highlighting the potential of miRNAs as diagnostic markers for ALS. miR-21 and miR-206 are aberrantly expressed in both mouse model and patient samples, positioning them as key potential diagnostic markers in ALS. Additionally, miR-29a, miR-29b, miR-181a, and miR-142-3p have shown aberrant expression in both types of samples and show promise as clinical targets for ALS. Finally, miR-1197 and miR-486b-5p have been recently identified as aberrantly expressed miRNAs in mouse models for ALS, although further studies are needed to determine their viability as diagnostic targets. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

Figure 1
<p>Morbidity and clinical manifestations of amyotrophic lateral sclerosis (ALS).</p>
Full article ">Figure 2
<p>Hypothesized pathogenesis of amyotrophic lateral sclerosis (ALS).</p>
Full article ">Figure 3
<p>Model of miRNA-promoted apoptosis. The long non-coding RNA NR3C2-8:1(lnc-NR3C) promotes p53-mediated apoptosis through the miR-129-5p/USP10 Axis. Adapted from Ref. [<a href="#B60-life-14-01125" class="html-bibr">60</a>] with permission from Springer Link.</p>
Full article ">Figure 4
<p>Molecular gene networks of ALS-related genes possibly modulated by miR-142-3p [<a href="#B48-life-14-01125" class="html-bibr">48</a>]. The 10 genes associated with ALS are presented as a gene network. The red color represents miRNA overexpression, which in this case is miR-142-3p. Black lines indicate miRNA/mRNA interactions. The orange line represents the regulation of a target gene, which has been validated by other research groups. The gray lines (solid and dashed) represent structural or functional relationships between genes or their encoded proteins. ALS, amyotrophic lateral sclerosis. Adapted from Ref. [<a href="#B47-life-14-01125" class="html-bibr">47</a>] with permission from Elsevier.</p>
Full article ">
27 pages, 1494 KiB  
Review
Integrating Mitochondrial Biology into Innovative Cell Therapies for Neurodegenerative Diseases
by Adaleiz Ore, James M. Angelastro and Cecilia Giulivi
Brain Sci. 2024, 14(9), 899; https://doi.org/10.3390/brainsci14090899 - 5 Sep 2024
Viewed by 502
Abstract
The role of mitochondria in neurodegenerative diseases is crucial, and recent developments have highlighted its significance in cell therapy. Mitochondrial dysfunction has been implicated in various neurodegenerative disorders, including Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis, and Huntington’s diseases. Understanding the impact of mitochondrial biology [...] Read more.
The role of mitochondria in neurodegenerative diseases is crucial, and recent developments have highlighted its significance in cell therapy. Mitochondrial dysfunction has been implicated in various neurodegenerative disorders, including Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis, and Huntington’s diseases. Understanding the impact of mitochondrial biology on these conditions can provide valuable insights for developing targeted cell therapies. This mini-review refocuses on mitochondria and emphasizes the potential of therapies leveraging mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, stem cell–derived secretions, and extracellular vesicles. Mesenchymal stem cell–mediated mitochondria transfer is highlighted for restoring mitochondrial health in cells with dysfunctional mitochondria. Additionally, attention is paid to gene-editing techniques such as mito-CRISPR, mitoTALENs, mito-ZNFs, and DdCBEs to ensure the safety and efficacy of stem cell treatments. Challenges and future directions are also discussed, including the possible tumorigenic effects of stem cells, off-target effects, disease targeting, immune rejection, and ethical issues. Full article
(This article belongs to the Special Issue Advances in Cell Therapy of Neurodegenerative Diseases)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic representation of mechanisms behind MSC-mediated mitochondrial transfer. MSCs can transfer healthy mitochondria to distressed cells, particularly those with dysfunctional mitochondria. Three main mechanisms for the intercellular transfer of whole mitochondria have been identified: tunneling nanotubes (TNTs), gap junctions, and certain types of extracellular vesicles. TNTs are membrane tubes that act as a bridge between more distant cells. Gap junctions are intercellular channels that form direct connections between nearby cells. EVs, lipid-based particles secreted by cells, may contain whole mitochondria or smaller mitochondrial fragments. It is important to note that not all EVs are large enough to contain functional mitochondria.</p>
Full article ">Figure 2
<p>Overview of mitochondrial gene-editing strategies with the potential use for neurodegeneration and other mtDNA-linked diseases.</p>
Full article ">Figure 3
<p>Mitochondria-derived vesicles (MDVs) are small, membrane-bound structures that separate from mitochondria and help maintain cellular balance and respond to stress. They assist in removing damaged mitochondrial components and preserving mitochondrial integrity and function, which are essential for overall cellular health and resilience against stress. MDVs play a significant role in maintaining cellular balance and mitochondrial quality control, which is crucial in neurodegenerative diseases associated with mitochondrial dysfunction and oxidative stress. TEM images (11,000×) were taken from wild-type mouse cortex, identifying mitochondrial protrusions and MDVs. Other experimental details can be found under [<a href="#B165-brainsci-14-00899" class="html-bibr">165</a>].</p>
Full article ">
13 pages, 1960 KiB  
Article
N-Terminal Fragments of TDP-43—In Vitro Analysis and Implication in the Pathophysiology of Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degeneration
by Anna A. Chami, Léa Bedja-Iacona, Elodie Richard, Debora Lanznaster, Sylviane Marouillat, Charlotte Veyrat-Durebex, Christian R. Andres, Philippe Corcia, Hélène Blasco and Patrick Vourc’h
Genes 2024, 15(9), 1157; https://doi.org/10.3390/genes15091157 - 1 Sep 2024
Viewed by 515
Abstract
Abnormal cytoplasmic aggregates containing the TDP-43 protein and its fragments are present in the central nervous system of the majority of patients with amyotrophic lateral sclerosis (ALS) and in patients with frontotemporal lobar degeneration (FTLD). Many studies have focused on the C-terminal cleavage [...] Read more.
Abnormal cytoplasmic aggregates containing the TDP-43 protein and its fragments are present in the central nervous system of the majority of patients with amyotrophic lateral sclerosis (ALS) and in patients with frontotemporal lobar degeneration (FTLD). Many studies have focused on the C-terminal cleavage products of TDP-43 (CTFs), but few have focused on the N-terminal products (NTFs), yet several works and their protein domain composition support the involvement of NTFs in pathophysiology. In the present study, we expressed six NTFs of TDP-43, normally generated in vivo by proteases or following the presence of pathogenic genetic truncating variants, in HEK-293T cells. The N-terminal domain (NTD) alone was not sufficient to produce aggregates. Fragments containing the NTD and all or part of the RRM1 domain produced nuclear aggregates without affecting cell viability. Only large fragments also containing the RRM2 domain, with or without the glycine-rich domain, produced cytoplasmic aggregates. Of these, only NTFs containing even a very short portion of the glycine-rich domain caused a reduction in cell viability. Our results provide insights into the involvement of different TDP-43 domains in the formation of nuclear or cytoplasmic aggregates and support the idea that work on the development of therapeutic molecules targeting TDP-43 must also take into account NTFs and, in particular, those containing even a small part of the glycine-rich domain. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Alignments of TDP-43 protein sequences in cleavage site regions. Alignments and sequence comparisons were performed using Seaview software.</p>
Full article ">Figure 2
<p>(<b>A</b>) Schematic representation (right) and 3D representation (left; obtained by AlphaFold) of the NTFs and the TDP-43WT protein. (<b>B</b>) RT-PCR analysis of expression of different constructs in HEK-293 cells after transfection (GFP and GAPDH primers for control). (<b>C</b>) Western blotting with antibody anti-N-terminal TDP-43 region (*) or Actin (control) on total proteins extracted from transfected HEK-293 cells. (<b>D</b>) Western blotting with antibody anti-GFP (*) or Actin (control) on total proteins extracted from transfected HEK-293 cells.</p>
Full article ">Figure 3
<p>(<b>A</b>) Immunocytochemical analysis of the expression of GFP alone, NTFs, and TDP-43 WT protein fused to GFP 48 h post-transfection of HEK-293T cells. Confocal microscopy; scale bar, 20 µm. (<b>B</b>) Western blot on total protein extracts (<b>C</b>, Crude), soluble fractions (SF), insoluble fractions (IF) of non-transfected (NT) HEK-293T cells, and cells transfected by plasmids expressing GFP alone, NTFs of TDP-43, and TDP-43 WT proteins fused to GFP. Antibody directed against the N-terminal region of TDP-43. Actin was used for internal control. (<b>C</b>) Cell viability analysis of cells transfected with an empty vector (TOPO) or with plasmids expressing GFP or TDP-43 NTFs or WT protein (n = 3, * <span class="html-italic">p</span> &lt; 0.05, One-Way, Dunnett’s test).</p>
Full article ">Figure 4
<p>(<b>A</b>) Schematic representation (right) and 3D representation (left; AlphaFold) of the TDP-43WT protein and NTF1-374. (<b>B</b>) Immunocytochemical analysis of the expression of TDP-43 WT and NTF1-374 proteins fused to GFP, 48 h post-transfection of HEK-293 cells. Fluorescent microscopy; scale bar, 20 μm. (<b>C</b>) Analysis of viability by trypan blue assay of HEK-293 cells transfected with an empty vector or plasmids expressing GFP or TDP-43 NTF1-374 or WT proteins (n = 3, * <span class="html-italic">p</span> &lt; 0.05, Kruskal–Wallis test).</p>
Full article ">
21 pages, 1511 KiB  
Review
Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases
by Mariagiovanna Di Chiano, Fabio Sallustio, Daniela Fiocco, Maria Teresa Rocchetti, Giuseppe Spano, Paola Pontrelli, Antonio Moschetta, Loreto Gesualdo, Raffaella Maria Gadaleta and Anna Gallone
Int. J. Mol. Sci. 2024, 25(17), 9489; https://doi.org/10.3390/ijms25179489 - 31 Aug 2024
Viewed by 690
Abstract
Neurodegenerative disorders are the main cause of cognitive and physical disabilities, affect millions of people worldwide, and their incidence is on the rise. Emerging evidence pinpoints a disturbance of the communication of the gut–brain axis, and in particular to gut microbial dysbiosis, as [...] Read more.
Neurodegenerative disorders are the main cause of cognitive and physical disabilities, affect millions of people worldwide, and their incidence is on the rise. Emerging evidence pinpoints a disturbance of the communication of the gut–brain axis, and in particular to gut microbial dysbiosis, as one of the contributors to the pathogenesis of these diseases. In fact, dysbiosis has been associated with neuro-inflammatory processes, hyperactivation of the neuronal immune system, impaired cognitive functions, aging, depression, sleeping disorders, and anxiety. With the rapid advance in metagenomics, metabolomics, and big data analysis, together with a multidisciplinary approach, a new horizon has just emerged in the fields of translational neurodegenerative disease. In fact, recent studies focusing on taxonomic profiling and leaky gut in the pathogenesis of neurodegenerative disorders are not only shedding light on an overlooked field but are also creating opportunities for biomarker discovery and development of new therapeutic and adjuvant strategies to treat these disorders. Lactiplantibacillus plantarum (LBP) strains are emerging as promising psychobiotics for the treatment of these diseases. In fact, LBP strains are able to promote eubiosis, increase the enrichment of bacteria producing beneficial metabolites such as short-chain fatty acids, boost the production of neurotransmitters, and support the homeostasis of the gut–brain axis. In this review, we summarize the current knowledge on the role of the gut microbiota in the pathogenesis of neurodegenerative disorders with a particular focus on the benefits of LBP strains in Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, autism, anxiety, and depression. Full article
(This article belongs to the Special Issue Molecular Insights into Neurotrophins and Neuropsychiatric Disorders)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Caption. Microbiota-gut-brain-axis. Both eubiosis and an intact gut barrier promote a physiological communication within the gut-brain axis. On the contrary, a dysbiosic GM is associated with a disrupted intestinal barrier and triggers the release of inflammatory mediators that, one reaching the brain, cause neuronal changes leading to the pathogenesis of neurodegenerative disorders.</p>
Full article ">Figure 2
<p>Caption. Dysbiosis affecting the gut-brain interaction. A Dysbalance of species abundance in the GM influences the gut-brain axis and plays a crucialrole in the pathogenesis of neurodegenerative disorders. Abbreviation: ALS amyotrophic lateral sclerosis, MS multiple sclerosis, AD Alzheimer disease, ASD autism spectrum disorder, PD Parkinson disease.</p>
Full article ">
21 pages, 1764 KiB  
Systematic Review
Redox Homeostasis, Gut Microbiota, and Epigenetics in Neurodegenerative Diseases: A Systematic Review
by Constantin Munteanu, Anca Irina Galaction, Marius Turnea, Corneliu Dan Blendea, Mariana Rotariu and Mădălina Poștaru
Antioxidants 2024, 13(9), 1062; https://doi.org/10.3390/antiox13091062 - 30 Aug 2024
Viewed by 486
Abstract
Neurodegenerative diseases encompass a spectrum of disorders marked by the progressive degeneration of the structure and function of the nervous system. These conditions, including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), Amyotrophic lateral sclerosis (ALS), and Multiple sclerosis (MS), often lead [...] Read more.
Neurodegenerative diseases encompass a spectrum of disorders marked by the progressive degeneration of the structure and function of the nervous system. These conditions, including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), Amyotrophic lateral sclerosis (ALS), and Multiple sclerosis (MS), often lead to severe cognitive and motor deficits. A critical component of neurodegenerative disease pathologies is the imbalance between pro-oxidant and antioxidant mechanisms, culminating in oxidative stress. The brain’s high oxygen consumption and lipid-rich environment make it particularly vulnerable to oxidative damage. Pro-oxidants such as reactive nitrogen species (RNS) and reactive oxygen species (ROS) are continuously generated during normal metabolism, counteracted by enzymatic and non-enzymatic antioxidant defenses. In neurodegenerative diseases, this balance is disrupted, leading to neuronal damage. This systematic review explores the roles of oxidative stress, gut microbiota, and epigenetic modifications in neurodegenerative diseases, aiming to elucidate the interplay between these factors and identify potential therapeutic strategies. We conducted a comprehensive search of articles published in 2024 across major databases, focusing on studies examining the relationships between redox homeostasis, gut microbiota, and epigenetic changes in neurodegeneration. A total of 161 studies were included, comprising clinical trials, observational studies, and experimental research. Our findings reveal that oxidative stress plays a central role in the pathogenesis of neurodegenerative diseases, with gut microbiota composition and epigenetic modifications significantly influencing redox balance. Specific bacterial taxa and epigenetic markers were identified as potential modulators of oxidative stress, suggesting novel avenues for therapeutic intervention. Moreover, recent evidence from human and animal studies supports the emerging concept of targeting redox homeostasis through microbiota and epigenetic therapies. Future research should focus on validating these targets in clinical settings and exploring the potential for personalized medicine strategies based on individual microbiota and epigenetic profiles. Full article
Show Figures

Figure 1

Figure 1
<p>The PRISMA flow diagram is used to illustrate the flow of information process [<a href="#B70-antioxidants-13-01062" class="html-bibr">70</a>].</p>
Full article ">Figure 2
<p>The interplay between diet, gut microbiota, epigenetic regulation, and oxidative stress in neurodegenerative diseases. The image depicts the gut–brain axis. It highlights the impact of diet, nutrients, and environmental agents on intestinal microbiota, shows oxidative stress pathways, and indicates the disruption of the blood–brain barrier (BBB). The figure highlights the role of gut-derived metabolites in modulating epigenetic mechanisms, such as DNA methylation and histone modifications. These epigenetic changes influence the expression of genes involved in inflammatory and oxidative stress responses. Furthermore, the figure depicts how dysbiosis, an imbalance in gut microbiota, can disrupt these protective mechanisms, leading to increased oxidative stress and aberrant epigenetic modifications. The figure was inspired by [<a href="#B157-antioxidants-13-01062" class="html-bibr">157</a>,<a href="#B158-antioxidants-13-01062" class="html-bibr">158</a>].</p>
Full article ">
Back to TopTop