bims-axbals Biomed News
on Axonal biology and ALS
Issue of 2025–04–20
twenty papers selected by
TJ Krzystek



  1. Int J Mol Sci. 2025 Mar 21. pii: 2867. [Epub ahead of print]26(7):
      Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons. Consequent to the loss of these cells, neuromuscular functions decline, causing progressive weakness, muscle wasting, and paralysis, leading to death in 2 to 5 years. More than 90% of ALS cases are sporadic, while the remaining 10% of cases are familial, due to mutations in 40 different genes. One of the most common genes to be mutated in ALS is TARDBP (transactive response DNA binding protein 43), which encodes TDP-43 (TAR DNA-binding protein 43). A mutation in exon 6 of TARDBP causes the aminoacidic substitution G376D in the C-terminal region of TDP-43, leading to its cytoplasmic mislocalization and aggregation. In fibroblasts derived from patients carrying this mutation, we found a strong increase in lysosome number, with overexpression and higher nuclear translocation of the transcription factor TFEB. In contrast, lysosomal functionality was deeply compromised. Interestingly, lysosomal activity was unaffected at an early stage of the disease, worsening in more advanced stages. Moreover, we observed the same pathological phenotype in iPSC (induced pluripotent stem cells)-derived patient motor neurons carrying the G376D mutation. Therefore, this mutation compromises the functionality of lysosomes, possibly contributing to neurodegeneration.
    Keywords:  TDP-43; TFEB; amyotrophic lateral sclerosis; lysosome; neurodegeneration; neurodegenerative disease
    DOI:  https://doi.org/10.3390/ijms26072867
  2. Methods Mol Biol. 2025 ;2910 53-68
      Reprogramming of somatic cells like blood cells and fibroblasts to obtain human induced pluripotent stem cells (hiPSCs) has become a state-of-the-art tool to study human diseases. The self-renewable hiPSCs offer ease of genetic modifications and can be differentiated into any cell type of the human body ranging from hepatocytes, cardiac myocytes, and subtypes of neurons. Dopaminergic (DA) neurons are one such neuronal subtype that is largely present in the midbrain region of the human brain and controls several functions like voluntary movement, reward, addiction, and stress. Loss of DA neurons is associated with one of the most common neurological disorders, Parkinson's disease (PD). Here, we describe a small molecule-directed approach for the generation of functionally mature dopaminergic neurons through the differentiation of hiPSCs. Differentiated DA neurons can be used to study their role in (patho)physiology.
    Keywords:  Astrocytes; Differentiation; Dopaminergic neurons; Induced pluripotent stem cells; Network activity; Patterning
    DOI:  https://doi.org/10.1007/978-1-0716-4446-1_4
  3. Transl Neurodegener. 2025 Apr 16. 14(1): 18
      Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
    Keywords:  Astrocytes; Frontotemporal dementia; Intercellular communication; Lipid dyshomeostasis; Microglia; Neurovasculature; Oligodendrocytes; Primary progressive aphasia; Progranulin deficiency; TAR-DNA binding protein 43
    DOI:  https://doi.org/10.1186/s40035-025-00475-8
  4. Transl Neurodegener. 2025 Apr 16. 14(1): 20
       BACKGROUND: Cytoplasmic aggregation of TAR DNA binding protein 43 (TDP-43) in neurons is one of the hallmarks of TDP-43 proteinopathy. Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are closely associated with TDP-43 proteinopathy; however, it remains uncertain whether TDP-43 aggregation initiates the pathology or is a consequence of it.
    METHODS: To demonstrate the pathology of TDP-43 aggregation, we applied the optoDroplet technique in Caenorhabditis elegans (C. elegans), which allows spatiotemporal modulation of TDP-43 phase separation and assembly.
    RESULTS: We demonstrate that optogenetically induced TDP-43 aggregates exhibited insolubility similar to that observed in TDP-43 proteinopathy. These aggregates increased the severity of neurodegeneration, particularly in GABAergic motor neurons, and exacerbated sensorimotor dysfunction in C. elegans.
    CONCLUSIONS: We present an optogenetic C. elegans model of TDP-43 proteinopathy that provides insight into the neuropathological mechanisms of TDP-43 aggregates. Our model serves as a promising tool for identifying therapeutic targets for TDP-43 proteinopathy.
    Keywords:   C. elegans ; Amyotrophic lateral sclerosis; Frontotemporal lobar degeneration; Neurodegenerative diseases; OptoDroplet; Optogenetics; TDP-43 proteinopathy
    DOI:  https://doi.org/10.1186/s40035-025-00480-x
  5. Methods Mol Biol. 2025 ;2910 27-36
      Generation of human induced pluripotent stem cells (iPSCs) provided a unique platform for human brain development studies, in vitro disease modeling, and therapeutic strategy development. Human stem cells can be rapidly and efficiently differentiated into several distinct subpopulations of brain cells. These stem cell-derived systems are gaining acceptance as a viable alternative in the neuroscience field as they can mimic interactions between various brain cells, and help recapitulate brain regions with specific functions. Here, we describe a method to generate functional, postmitotic, excitatory cortical-like neurons from iPSCs by expressing the NGN2 transgene from a stably integrated doxycycline-inducible promoter. These induced neurons (iNs) can be utilized to study the development and function of human cortical neurons. They also allow studying disease mechanisms by comparing normal and pathophysiological conditions and enable reliable screens for testing of therapeutic approaches.
    Keywords:  Differentiation; Human neurons; NGN2; Stem cells; Synapse; iPSCs
    DOI:  https://doi.org/10.1007/978-1-0716-4446-1_2
  6. Methods Mol Biol. 2025 ;2910 105-125
      The expression pattern of proteins defines the range of biological processes in cellular subcompartments. A core aim in cell biology is therefore to determine the localization and composition of protein complexes within cells. Proximity labeling methodologies offer an unbiased and efficient way to unravel the cellular micro-environment of proteins, providing insights into the molecular networks they participate in. In this chapter, we present a protocol for conducting proximity labeling experiments in primary murine neuronal cultures in vitro based on the proximity-dependent biotinylation identification (BioID) approach. Data acquired through this protocol can be utilized to identify the composition of protein complexes in neurons and to create molecular maps of neuronal subcompartments. This will aid in determining the spatial distribution of biological processes within neurons, and in unraveling fundamental principles of neuronal function and plasticity.
    Keywords:  Affinity purification; Biotinylation identification (BioID); Mass spectrometry; Primary neurons; Proximity labeling; Synapses; TurboID
    DOI:  https://doi.org/10.1007/978-1-0716-4446-1_7
  7. Development. 2025 Apr 14. pii: dev.204424. [Epub ahead of print]
      During neuronal development, microtubule reorganization shapes axons and dendrites, establishing the framework for efficient nervous system wiring. Our previous work demonstrated the role of kinesin-1 in driving microtubule sliding, which powers early axon outgrowth and regeneration in Drosophila melanogaster. Here, we reveal a critical new role of kinesin-5, a mitotic motor, in modulating postmitotic neuron development. The Drosophila kinesin-5, Klp61F, is expressed in larval brain neurons, with high levels in ventral nerve cord (VNC) neurons. Knockdown of Klp61F in neurons leads to severe adult locomotion defects and lethality, primarily due to defects in VNC motor neurons. Klp61F depletion results in excessive microtubule penetration into the axon growth cone, causing significant axon growth defects in culture and in vivo. These defects are rescued by a chimeric human-Drosophila kinesin-5 motor, indicating a conserved role of kinesin-5 in neuronal development. Altogether, we propose that kinesin-5 acts as a brake on kinesin-1-driven microtubule sliding, ensuring proper axon pathfinding in growing neurons.
    Keywords:  Axonal growth; Kinesin-5; Microtubules; Mitotic motor; Motor neurons; Neuronal development
    DOI:  https://doi.org/10.1242/dev.204424
  8. NPJ Syst Biol Appl. 2025 Apr 17. 11(1): 37
      MIRO1 is a mitochondrial outer membrane protein important for mitochondrial distribution, dynamics and bioenergetics. Over the last decade, evidence has pointed to a link between MIRO1 and Parkinson's disease (PD) pathogenesis. Moreover, a heterozygous MIRO1 mutation (p.R272Q) was identified in a PD patient, from which an iPSC-derived midbrain organoid model was derived, showing MIRO1 mutant-dependent selective loss of dopaminergic neurons. Herein, we use patient-specific iPSC-derived midbrain organoids carrying the MIRO1 p.R272Q mutation to further explore the cellular and molecular mechanisms involved in dopaminergic neuron degeneration. Using single-cell RNA sequencing (scRNAseq) analysis and metabolic modeling we show that the MIRO1 p.R272Q mutation affects the dopaminergic neuron developmental path leading to metabolic deficits and disrupted neuron-astrocyte metabolic crosstalk, which might represent an important pathogenic mechanism leading to their loss.
    DOI:  https://doi.org/10.1038/s41540-025-00509-x
  9. J Comput Neurosci. 2025 Apr 17.
      Synaptic and neural properties can change during periods of auditory deprivation. These changes may disrupt the computations that neurons perform. In the brainstem of chickens, auditory deprivation can lead to changes in the size and biophysics of the axon initial segment (AIS) of neurons in the sound source localization circuit. This is the phenomenon of axon initial segment (AIS) plasticity. Individuals who use cochlear implants (CIs) experience periods of hearing loss, and so we ask whether AIS plasticity in neurons of the medial superior olive (MSO), a key stage of sound location processing, would impact time difference sensitivity in the scenario of hearing with cochlear implants. The biophysical changes that we implement in our model of AIS plasticity include enlargement of the AIS and replacement of low-threshold potassium conductance with the more slowly-activated M-type potassium conductance. AIS plasticity has been observed to have a homeostatic effect with respect to excitability. In our model, AIS plasticity has the additional effect of converting MSO neurons from phasic firing type to tonic firing type. Phasic firing is known to have greater temporal sensitivity to coincident inputs. Consistent with this, we find AIS plasticity degrades time difference sensitivity in the auditory deprived MSO neuron model across a range of stimulus parameters. Our study illustrates a possible mechanism of cellular plasticity in a non-peripheral stage of neural processing that could impose barriers to sound source localization by bilateral cochlear implant users.
    Keywords:  Axon initial segment; Cochlear implant; Computer model; Medial superior olive; Plasticity; Sound localization
    DOI:  https://doi.org/10.1007/s10827-025-00902-9
  10. Sci Rep. 2025 Apr 12. 15(1): 12606
      The widespread protozoan Toxoplasma gondii chronically infects neural tissue in vertebrates and is linked to various neurological and neuropsychiatric disorders in humans. However, its effects on sparsely infected neurons and on broader neural circuits remain elusive. Our study reveals that T. gondii infection disrupts cytoskeletal dynamics in SH-SY5Y neuronal cells and primary cortical neurons. Infected neuronal cells undergo significant cytomorphological changes, including retraction of dendritic extensions and alterations in microtubule and F-actin networks, across both parasite genotypes I and II. These cytoskeletal alterations were notably diminished in cells exposed to T. gondii mutants with impaired secretion via the MYR translocon, and were independent of intraneuronal parasite replication. Moreover, a bystander effect was observed, with supernatants from T. gondii-challenged cells inducing similar cytoskeletal changes in uninfected cells. Analyses of extracellular vesicles (EVs) in supernatants revealed differential expression of host microRNAs in response to infection, most notably the upregulation of miR-221-3p, a microRNA not previously associated with T. gondii. The data indicate that unidentified parasite-derived effector(s) secreted via the MYR translocon, in conjunction with MYR-independently induced EV-associated host microRNAs, mediate cytoskeletal alterations in both infected and bystander neuronal cells. The findings provide new insights into molecular mechanisms by which T. gondii infection may disrupt neural networks, shedding light on its potential role in neuronal dysregulation.
    Keywords:  Apicomplexan parasites; Cytoskeleton; Extracellular vesicle; Host–pathogen interaction; Neuron; microRNA
    DOI:  https://doi.org/10.1038/s41598-025-96298-8
  11. Nat Commun. 2025 Apr 17. 16(1): 3641
      Biogenesis of mitoribosomes requires dedicated chaperones, RNA-modifying enzymes, and GTPases, and defects in mitoribosome assembly lead to severe mitochondriopathies in humans. Here, we characterize late-step assembly states of the small mitoribosomal subunit (mtSSU) by combining genetic perturbation and mutagenesis analysis with biochemical and structural approaches. Isolation of native mtSSU biogenesis intermediates via a FLAG-tagged variant of the GTPase MTG3 reveals three distinct assembly states, which show how factors cooperate to mature the 12S rRNA. In addition, we observe four distinct primed initiation mtSSU states with an incompletely matured rRNA, suggesting that biogenesis and translation initiation are not mutually exclusive processes but can occur simultaneously. Together, these results provide insights into mtSSU biogenesis and suggest a functional coupling between ribosome biogenesis and translation initiation in human mitochondria.
    DOI:  https://doi.org/10.1038/s41467-025-58827-x
  12. Trends Biotechnol. 2025 Apr 11. pii: S0167-7799(25)00046-0. [Epub ahead of print]
      Brain organoids are 3D tissue models of the human brain that are derived from pluripotent stem cells (PSCs). They have enabled studies that were previously stymied by the inaccessibility of human brain tissue or the limitations of mouse models of some brain diseases. Despite their enormous potential, brain organoids have had significant limitations that prevented them from recapitulating the full complexity of the human brain and reduced their utility in disease studies. We describe recent progress in addressing these limitations, especially building complex organoids that recapitulate the interactions between multiple brain regions, and reconstructing in vitro the neural circuitry present in in vivo. These major advances in the human brain organoid technology will remarkably facilitate brain disease modeling and neuroscience research.
    Keywords:  assembloids; brain development; brain disease; brain organoids; neural circuitry
    DOI:  https://doi.org/10.1016/j.tibtech.2025.02.009
  13. Traffic. 2025 Apr;26(4-6): e70007
      In certain kinds of cells, clathrin-independently endocytosed cargo proteins are recycled back to the plasma membrane via specialized tubular-shaped endosomes, so-called tubular endosomes. Several regulators, including Rab small GTPases, have previously been reported to control tubular endosome structures, and one of the regulators, Rab22A, controls cargo sorting and tubule elongation. Since Rab activity is generally controlled by a guanine nucleotide exchange factor (GEF) and a GTPase-activating protein (GAP), these upstream regulators would also be involved in tubular endosome formation. However, although we have previously reported that Vps9d1 is a Rab22A-GEF that controls tubular endosome formation, there have been no reports of Rab-GAPs that are required for tubular endosome formation. Here, we demonstrated by comprehensive screening of TBC/Rab-GAPs that four Rab-GAPs, TBC1D10B, TBC1D18, TBC1D22B and EVI5, are involved in tubular endosome formation in HeLa cells in a GAP-activity-dependent manner. Knockdown or overexpression of each of these Rab-GAPs resulted in the same phenotype, that is, reduced tubular endosome structures. Since one of these four Rab-GAPs, TBC1D10B, was able to reduce the amount of active Rab22A and the size of Rab22A-positive early endosomes, it is the most probable candidate for a Rab22A-GAP. Our findings suggest that a proper GTPase cycle is important for the control of tubular endosome formation.
    Keywords:  GTPase‐activating protein (GAP); Rab22A; Tre‐2/Bub2/Cdc16 (TBC) domain; comprehensive screening; membrane traffic; small GTPase; tubular endosome
    DOI:  https://doi.org/10.1111/tra.70007
  14. Int J Mol Sci. 2025 Apr 01. pii: 3263. [Epub ahead of print]26(7):
      Retinal degenerative diseases (RDDs) comprise diverse genetic and phenotypic conditions that cause progressive retinal dysfunction and cell loss, leading to vision impairment or blindness. Most RDDs lack appropriate animal models for their study, which affects understanding their disease mechanisms and delays the progress of new treatment development. Recent advances in stem cell engineering, omics, and organoid technology are facilitating research into diseases for which there are no previously existing models. The development of retinal organoids produced from human stem cells has impacted the study of retinal development as well as the development of in vitro models of diseases, opening possibilities for applications in regenerative medicine, drug discovery, and precision medicine. In this review, we recapitulate research in the retinal organoid models for RDD, mentioning some of the main pathways underlying retinal neurodegeneration that can be studied in these new models, as well as their limitations and future challenges in this rapidly advancing field.
    Keywords:  photoreceptors; retinal degeneration; retinal organoids; stem cells
    DOI:  https://doi.org/10.3390/ijms26073263
  15. J Mol Biol. 2025 Apr 10. pii: S0022-2836(25)00204-9. [Epub ahead of print] 169138
      Autophagy is a conserved cellular recycling pathway that delivers damaged or superfluous cytoplasmic material to lysosomes for degradation. In response to cytotoxic stress or starvation, autophagy can also sequester bulk cytoplasm and deliver it to lysosomes to regenerate building blocks. In macroautophagy, a membrane cisterna termed phagophore that encloses autophagic cargo is generated. The formation of the phagophore depends on a conserved machinery of autophagy related proteins. The phosphatidylinositol(3)-phosphate binding protein WIPI2 facilitates the transition from phagophore initiation to phagophore expansion by recruiting the ATG12-ATG5-ATG16L1 complex to phagophores. This complex functions as an E3-ligase to conjugate ubiquitin-like ATG8 proteins to phagophore membranes, which promotes tethering of cargo to phagophore membranes, phagophore expansion, maturation and the fusion of autophagosomes with lysosomes. ATG16L1 also has important functions independently of ATG12-ATG5 in autophagy and beyond. In this review, we will summarize the functions of WIPI2 and ATG16L1 in selective and nonselective autophagy.
    DOI:  https://doi.org/10.1016/j.jmb.2025.169138
  16. Trends Mol Med. 2025 Apr 14. pii: S1471-4914(25)00059-0. [Epub ahead of print]
      Phosphoglycerate kinase 1 (PGK1) is a highly conserved enzyme that catalyzes the initial ATP-producing step in glycolysis. Improving cellular energy production by increasing PGK1 activity may be beneficial in multiple neurological conditions where cell metabolism is dysregulated, including Parkinson's disease (PD) and motor neuron disease (MND). This review examines recent evidence that suggests increasing PGK1 activity may be beneficial in multiple neurological conditions and discusses the current challenges surrounding the development of PGK1-focused therapies. PGK1 has considerable therapeutic potential, but novel PGK1 activators are needed to maximize the benefit for patients.
    Keywords:  amyotrophic lateral sclerosis; neurodegeneration; spinal muscular atrophy; terazosin; therapy development
    DOI:  https://doi.org/10.1016/j.molmed.2025.03.008
  17. J Tissue Eng Regen Med. 2023 ;2023 9802235
      Healthy skeletal muscle can regenerate after ischaemic, mechanical, or toxin-induced injury, but ageing impairs that regeneration potential. This has been largely attributed to dysfunctional satellite cells and reduced myogenic capacity. Understanding which signalling pathways are associated with reduced myogenesis and impaired muscle regeneration can provide valuable information about the mechanisms driving muscle ageing and prompt the development of new therapies. To investigate this, we developed a high-throughput in vitro model to assess muscle regeneration in chemically injured C2C12 and human myotube-derived young and aged myoblast cultures. We observed a reduced regeneration capacity of aged cells, as indicated by an attenuated recovery towards preinjury myotube size and myogenic fusion index at the end of the regeneration period, in comparison with younger muscle cells that were fully recovered. RNA-sequencing data showed significant enrichment of KEGG signalling pathways, PI3K-Akt, and downregulation of GO processes associated with muscle development, differentiation, and contraction in aged but not in young muscle cells. Data presented here suggest that repair in response to in vitro injury is impaired in aged vs. young muscle cells. Our study establishes a framework that enables further understanding of the factors underlying impaired muscle regeneration in older age.
    DOI:  https://doi.org/10.1155/2023/9802235
  18. Mol Cell. 2025 Apr 17. pii: S1097-2765(25)00196-0. [Epub ahead of print]85(8): 1487-1508
      Mitochondria serve as central hubs in cellular metabolism by sensing, integrating, and responding to metabolic demands. This integrative function is achieved through inter-organellar communication, involving the exchange of metabolites, lipids, and signaling molecules. The functional diversity of metabolite exchange and pathway interactions is enabled by compartmentalization within organelle membranes. Membrane contact sites (MCSs) are critical for facilitating mitochondria-organelle communication, creating specialized microdomains that enhance the efficiency of metabolite and lipid exchange. MCS dynamics, regulated by tethering proteins, adapt to changing cellular conditions. Dysregulation of mitochondrial-organelle interactions at MCSs is increasingly recognized as a contributing factor in the pathogenesis of multiple diseases. Emerging technologies, such as advanced microscopy, biosensors, chemical-biology tools, and functional genomics, are revolutionizing our understanding of inter-organellar communication. These approaches provide novel insights into the role of these interactions in both normal cellular physiology and disease states. This review will highlight the roles of metabolite transporters, lipid-transfer proteins, and mitochondria-organelle interfaces in the coordination of metabolism and transport.
    Keywords:  endoplasmic reticulum; inter-organellar communication; mitochondria; organellar metabolism; organelle membrane contact sites
    DOI:  https://doi.org/10.1016/j.molcel.2025.03.003
  19. Redox Biol. 2025 Apr 03. pii: S2213-2317(25)00139-9. [Epub ahead of print]83 103626
      Many life processes are regulated by physiological redox signaling, but excessive oxidative stress can damage biomolecules and contribute to disease. Neuronal microtubules are critically involved in axon homeostasis, regulation of axonal transport, and neurodegenerative processes. However, whether and how physiological redox signaling affects axonal microtubules is largely unknown. Using live cell imaging and super-resolution microscopy, we show that subtoxic concentrations of the central redox metabolite hydrogen peroxide increase axonal microtubule dynamics, alter the structure of the axonal microtubule array, and affect the efficiency of axonal transport. We report that the mitochondria-targeting antioxidant SkQ1 and the microtubule stabilizer EpoD abolish the increase in microtubule dynamics. We found that hydrogen peroxide specifically modulates the phosphorylation state of microtubule-regulating proteins, which differs from arsenite as an alternative stress inducer, and induces a largely non-overlapping phosphorylation pattern of MAP1B as a main target. Cell-wide phosphoproteome analysis revealed signaling pathways that are inversely activated by hydrogen peroxide and arsenite. In particular, hydrogen peroxide treatment was associated with kinases that suppress apoptosis and regulate brain metabolism (PRKDC, CK2, PDKs), suggesting that these pathways play a central role in physiological redox signaling and modulation of axonal microtubule organization. The results suggest that the redox metabolite and second messenger hydrogen peroxide induces rapid and local reorganization of the microtubule array in response to mitochondrial activity or as a messenger from neighboring cells by activating specific signaling cascades.
    Keywords:  Axon; Hydrogen peroxide; Microtubules; Redox signalling; Tau
    DOI:  https://doi.org/10.1016/j.redox.2025.103626
  20. J Neural Transm (Vienna). 2025 Apr 17.
      Although α-synuclein pathology is typically associated with Lewy body diseases and multiple systems atrophy, increasing evidence indicates that it also occurs in a group of lysosomal storage disorders termed sphingolipidoses caused by the incomplete degradation, and subsequent accumulation, of a class of lipids termed sphingolipids. Notably, a number of genes that cause sphingolipidoses are also risk genes for Lewy body diseases, suggesting aetiological links between these distinct disorders. In the present review, we discuss the sphingolipidoses in which α-synuclein pathology has been reported: Gaucher disease, Krabbe disease, metachromatic leukodystrophy, Tay-Sachs disease and Anderson-Fabry disease, and describe the characteristic clinical and pathological features of these disorders, in addition to the evidence suggesting α-synuclein pathology occurs in these disorders. Finally, we evaluate the pathological mechanisms that underlie these rare disorders, with particular attention to how the enzymatic deficiency, substrate accumulation, or both, could contribute to the genesis of α-synuclein pathology and the implications of this for Lewy body diseases.
    Keywords:  Alpha-synuclein; Lewy; Lysosomal storage disorder; Neurodegeneration; Parkinson’s; Sphingolipid
    DOI:  https://doi.org/10.1007/s00702-025-02925-z