1
|
Ishii M, Tsurusaki A, Komatsu M, Shiozaki K. Neu1-deficient zebrafish cells exhibit reduced Edwardsiella piscicida infection due to altered lysosomal exocytosis and membrane dynamics. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110273. [PMID: 40074189 DOI: 10.1016/j.fsi.2025.110273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/14/2025]
Abstract
Edwardsiella piscicida is a Gram-negative intracellular pathogen causing Edwardsiellosis, leading to economic losses in aquaculture. While phagocytosis is its primary infection route, alternative entry pathways remain largely unexplored. Neu1 sialidase, a lysosomal enzyme in glycoconjugate degradation, was investigated for its role in E. piscicida infection using primary cultured cells derived from Neu1-KO zebrafish fin (Neu1-KO cells). Compared to wild-type (WT) cells, Neu1-KO cells exhibited lower infection rates, which were associated with enhanced lysosomal exocytosis. Infection was restored by the intracellular calcium chelator BAPTA-AM, highlighting the role of exocytosis. Leupeptin, a cysteine/serine protease inhibitor, increased E. piscicida infection in Neu1-KO cells. Neu1-KO cells exhibited lower rab10 expression and reduced membrane ruffling, which was restored by BAPTA-AM and leupeptin. Given the role of epidermal growth factor receptor (EGFR) signaling, we assessed its phosphorylation, which was reduced in Neu1-KO cells but restored by treatment with BAPTA-AM and leupeptin. This suggests that inhibiting lysosome exocytosis or extracellular protease activity may enhance EGFR phosphorylation. These findings indicate that the decreased E. piscicida infection in Neu1-KO cells resulted from enhanced lysosomal exocytosis, leading to increased extracellular protease secretion, subsequent EGFR inactivation by extracellular protease, and reduced EGFR-regulated ruffling. This study provides novel insights into the regulatory mechanisms of bacterial infection and lysosomal exocytosis, informing potential therapeutic strategies against intracellular pathogens.
Collapse
Affiliation(s)
- Mika Ishii
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, 890-0056, Japan
| | - Akari Tsurusaki
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, 890-0056, Japan
| | - Masaharu Komatsu
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, 890-0056, Japan; Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Kazuhiro Shiozaki
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, 890-0056, Japan; Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan.
| |
Collapse
|
2
|
Viana GM, Pan X, Fan S, Xu T, Wyatt A, Pshezhetsky AV. Cathepsin B inhibition blocks amyloidogenesis in the mouse models of neurological lysosomal diseases MPS IIIC and sialidosis. Mol Ther Methods Clin Dev 2025; 33:101432. [PMID: 40092638 PMCID: PMC11910108 DOI: 10.1016/j.omtm.2025.101432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Neuronal accumulation of amyloid aggregates is a hallmark of brain pathology in neurological lysosomal storage diseases (LSDs), including mucopolysaccharidoses (MPS); however, the molecular mechanism underlying this pathology has not been understood. We demonstrate that elevated lysosomal cathepsin B (CTSB) levels and CTSB leakage to the cytoplasm triggers amyloidogenesis in two neurological LSDs. CTSB levels were elevated 3- to 5-fold in the cortices of mouse models of MPS IIIC (Hgsnat-Geo and Hgsnat P304L ) and sialidosis (Neu1 ΔEx3 ), as well as in cortical samples of MPS I, IIIA, IIIC, and IIID patients. CTSB was found in the cytoplasm of pyramidal layer IV-V cortical neurons containing thioflavin-S+, β-amyloid+ aggregates consistent with a pro-senile phenotype. In contrast, CTSB-deficient MPS IIIC (Hgsnat P304L /Ctsb -/- ) mice as well as Hgsnat P304L and Neu1 ΔEx3 mice chronically treated with irreversible brain-penetrable CTSB inhibitor E64 showed a drastic reduction in neuronal thioflavin-S+/APP+ deposits. Neurons of Hgsnat P304L /Ctsb -/- mice and E64-treated Hgsnat P304L mice also showed reduced levels of P62+, LC3+ puncta, GM2 ganglioside, and misfolded subunit C of mitochondrial ATP synthase, consistent with restored autophagy. E64 treatment also rescued hyperactivity and reduced anxiety in Hgsnat P304L mice, implying that CTSB may become a novel pharmacological target for MPS III and similar LSDs.
Collapse
Affiliation(s)
- Gustavo M Viana
- Division of Medical Genetics, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Centre, Montreal, QC H3A 0C7, Canada
| | - Xuefang Pan
- Division of Medical Genetics, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Centre, Montreal, QC H3A 0C7, Canada
| | - Shuxian Fan
- Division of Medical Genetics, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Centre, Montreal, QC H3A 0C7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3T 1C5, Canada
| | - TianMeng Xu
- Division of Medical Genetics, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Centre, Montreal, QC H3A 0C7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3T 1C5, Canada
| | - Alexandra Wyatt
- Division of Medical Genetics, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Centre, Montreal, QC H3A 0C7, Canada
| | - Alexey V Pshezhetsky
- Division of Medical Genetics, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Centre, Montreal, QC H3A 0C7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3T 1C5, Canada
| |
Collapse
|
3
|
Kılıç M, İcil S, Sezer A, Kaya-Güneş Ö, Comoğlu SS. Sialidosis type 1 in a Turkish family: a case report and review of literatures. J Pediatr Endocrinol Metab 2025; 38:176-186. [PMID: 39733340 DOI: 10.1515/jpem-2024-0468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024]
Abstract
OBJECTIVES Sialidosis type 1 is a rare autosomal recessive lysosomal storage disorder caused by pathogenic variants in the NEU1 gene, which encodes the sialic acid-degrading enzyme α-neuraminidase. Sialidosis type 1 is a milder form with a late-onset phenotype, characterized by progressive myoclonic epilepsy and ataxia with cherry-red spots. Sialidosis type 2 is an early-onset and more severe form presenting with dysmorphic features, hepatosplenomegaly and cognitive delay. Clinical diagnosis is usually supported by increased urinary bound sialic acid excretion and confirmed by genetic analysis or demonstration of α-neuraminidase enzyme deficiency in cultured fibroblasts. The aim of this study was to present a case of type 1 sialidosis, review the literature, and investigate genotype-phenotype correlations, symptom frequencies, and race-specific mutations in patients diagnosed with type 1 sialidosis. CASE PRESENTATION We report herein a family of four Turkish siblings affected with sialidosis type 1 associated with a homozygous variant, c.403G>A p. (Asp135Asn), in the NEU1 gene. A systematic literature review on sialidosis type 1 was carried out, by the PubMed database was searched using keywords included sialidosis and/or NEU1 gene. We selected case reports or series that included genetically confirmed type 1 sialidosis from 1996 to 2023. So far, nearly genetically confirmed 80 patients from unrelated 65 families, more than 40 NEU1 disease causing mutations, have been identified in patients with sialidosis type 1. Among the reported mutations, missense variants are the most common, and few nonsense, frameshift, exonic duplications or small deletions have been reported. c.239C>T p. (Pro80Leu) variant in Chinese and Japanese patients, c.649G>A p. (Val217Met) variant in Japanese patients, c.880C>T p. (Arg294Cys) variant in Indian patients, c.629C>T p. (Pro210Leu) variant in Ecuadorian patients, c.982G>A p. (Gly328Ser) variant in Italian patients, and c.403G>A p (Asp135Asn) and c.625del p. (Glu209Serfs*94) variants in Turkish patients were found higher. CONCLUSIONS Race-specific variants were found with higher percentages in certain populations.
Collapse
Affiliation(s)
- Mustafa Kılıç
- Department of Pediatrics, Metabolism Unit, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Suzan İcil
- Department of Pediatrics, Metabolism Unit, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Abdullah Sezer
- Department of Genetics, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Öznur Kaya-Güneş
- Department of Genetics, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Selim S Comoğlu
- Department of Neurology, Ankara Etlik City Hospital, Ankara, Türkiye
| |
Collapse
|
4
|
Fremuth LE, Hu H, van de Vlekkert D, Annunziata I, Weesner JA, Alessandra d'Azzo. Neuraminidase 1 regulates neuropathogenesis by governing the cellular state of microglia via modulation of Trem2 sialylation. Cell Rep 2025; 44:115204. [PMID: 39817909 PMCID: PMC11874873 DOI: 10.1016/j.celrep.2024.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
Neuraminidase 1 (NEU1) cleaves terminal sialic acids from sialoglycoproteins in endolysosomes and at the plasma membrane. As such, NEU1 regulates immune cells, primarily those of the monocytic lineage. Here, we examine how Neu1 influences microglia by modulating the sialylation of full-length Trem2 (Trem2-FL), a multifunctional receptor that regulates microglial survival, phagocytosis, and cytokine production. When Neu1 is deficient/downregulated, Trem2-FL remains sialylated, accumulates intracellularly, and is excessively cleaved into a C-terminal fragment (Trem2-CTF) and an extracellular soluble domain (sTrem2), enhancing their signaling capacities. Sialylated Trem2-FL (Sia-Trem2-FL) does not hinder Trem2-FL-DAP12-Syk complex assembly but impairs signal transduction through Syk, ultimately abolishing Trem2-dependent phagocytosis. Concurrently, Trem2-CTF-DAP12 complexes dampen NF-κB signaling, while sTrem2 propagates Akt-dependent cell survival and NFAT1-mediated production of TNF-α and CCL3. Because NEU1 and Trem2 are implicated in neurodegenerative/neuroinflammatory diseases, including Alzheimer disease and sialidosis, modulating NEU1 activity represents a therapeutic approach to broadly regulate microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Leigh Ellen Fremuth
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Huimin Hu
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Ida Annunziata
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Compliance Office, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jason Andrew Weesner
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
5
|
Monkhouse H, Deane JE. Linking glycosphingolipid metabolism to disease-related changes in the plasma membrane proteome. Biochem Soc Trans 2024; 52:2477-2486. [PMID: 39641585 DOI: 10.1042/bst20240315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Glycosphingolipids (GSLs) are vital components of the plasma membrane (PM), where they play crucial roles in cell function. GSLs form specialised membrane microdomains that organise lipids and proteins into functional platforms for cell adhesion and signalling. GSLs can also influence the function of membrane proteins and receptors, via direct protein-lipid interactions thereby affecting cell differentiation, proliferation, and apoptosis. Research into GSL-related diseases has primarily focussed on lysosomal storage disorders, where defective enzymes lead to the accumulation of GSLs within lysosomes, causing cellular dysfunction and disease. However, recent studies are uncovering the broader cellular impact of GSL imbalances including on a range of organelles and cellular compartments such as the mitochondria, endoplasmic reticulum and PM. In this review we describe the mechanisms by which GSL imbalances can influence the PM protein composition and explore examples of the changes that have been observed in the PM proteome upon GSL metabolic disruption. Identifying and understanding these changes to the PM protein composition will enable a more complete understanding of lysosomal storage diseases and provide new insights into the pathogenesis of other GSL-related diseases, including cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Holly Monkhouse
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, U.K
| | - Janet E Deane
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, U.K
| |
Collapse
|
6
|
Ding Y, Cheng M, Gong C. Two cases of type I sialidosis and a literature review. Orphanet J Rare Dis 2024; 19:440. [PMID: 39605025 PMCID: PMC11600752 DOI: 10.1186/s13023-024-03431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE This study aims to compare the clinical and electrophysiological characteristics of two cases of type I sialidosis in Chinese children with those reported in prior literature. The goal is to elucidate the clinical and genetic features of type I sialidosis. METHODS Clinical investigations and genetic analyses were conducted on an 11-year-old girl, primarily presenting with short stature, who was admitted in June 2020, and a 10-year-old boy, admitted in July 2023, exhibiting rapid weight gain and accompanying visual impairment as primary manifestations. A literature review was performed by summarizing data from 31 published articles encompassing 69 genetically confirmed cases of type I sialidosis up to 2023 for comparative analysis. RESULTS Patient 1 exhibited short stature, self-reported poor night vision, a history of occasional febrile seizures, mild scoliosis, bilateral cherry-red spots in the fundus, and prolonged P100 latency in both eyes as observed in visual evoked potentials (VEP). Genetic analysis revealed that she carried compound-heterozygous variants c.239 C > T (p.P80L) and c.880 C > T (p.R294C) in the NEU1 gene, inherited from her parents. Patient 2 presented with rapid weight gain and visual impairment, bilateral cherry-red spots in the fundus, abnormal neuroepithelial layer reflexes in both macular areas, approximately normal P100 latency but severely reduced amplitude in VEP after pupillary dilation, and severe bilateral optic nerve conduction block with relatively normal retinal cell function. Compound-heterozygous variants c.239 C > T (p.P80L) and c.803 A > G (p.T268C) were identified in the NEU1 gene of the Patient 2, inherited from his parents. By combining the cases reported in 31 literature articles with the 2 cases in our study, a total of 71 type I sialidosis patients were analyzed. The most common symptoms observed were muscle spasms (91.5%), followed by ataxia (75%) and seizures (63.6%). Intellectual impairment and abnormal electroencephalograms were more prevalent in Caucasian patients. Additionally, abnormal somatosensory evoked potentials, large cortical waves, and prolonged latency of VEP were more frequently observed in both Asian and Caucasian patients, serving as alternative indicators for early diagnosis. CONCLUSION NEU1 gene analysis provides essential guidance for genetic counseling and prenatal diagnosis. The exon 2 variant c.239 C > T (p.P80L) in the NEU1 gene may represent a mutation hotspot among Chinese patients.
Collapse
Affiliation(s)
- Yuan Ding
- Department of Endocrinology, Beijing Children's Hospital, Capital Medical University, National Centre for Children's Health, Genetics, Metabolism, Beijing, 100045, China
- MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56#Nan Lishi Rd, West District, Beijing, 100045, China
| | - Ming Cheng
- Department of Endocrinology, Beijing Children's Hospital, Capital Medical University, National Centre for Children's Health, Genetics, Metabolism, Beijing, 100045, China
- MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56#Nan Lishi Rd, West District, Beijing, 100045, China
| | - Chunxiu Gong
- Department of Endocrinology, Beijing Children's Hospital, Capital Medical University, National Centre for Children's Health, Genetics, Metabolism, Beijing, 100045, China.
- MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56#Nan Lishi Rd, West District, Beijing, 100045, China.
| |
Collapse
|
7
|
Benitez BA, Wallace CE, Patel M, Nykanen NP, Yuede CM, Eaton SL, Pottier C, Cetin A, Johnson M, Bevan MT, Gardiner WD, Edwards HM, Doherty BM, Harrigan RT, Kurian D, Wishart TM, Smith C, Cirrito JR, Sands MS. Haploinsufficiency of lysosomal enzyme genes in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.16.623962. [PMID: 39605615 PMCID: PMC11601326 DOI: 10.1101/2024.11.16.623962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
There is growing evidence suggesting that the lysosome or lysosome dysfunction is associated with Alzheimer's disease (AD). Pathway analysis of post mortem brain-derived proteomic data from AD patients shows that the lysosomal system is perturbed relative to similarly aged unaffected controls. However, it is unclear if these changes contributed to the pathogenesis or are a response to the disease. Consistent with the hypothesis that lysosome dysfunction contributes to AD pathogenesis, whole genome sequencing data indicate that heterozygous pathogenic mutations and predicted protein-damaging variants in multiple lysosomal enzyme genes are enriched in AD patients compared to matched controls. Heterozygous loss-of-function mutations in the palmitoyl protein thioesterase-1 (PPT1), α-L-iduronidase (IDUA), β-glucuronidase (GUSB), N-acetylglucosaminidase (NAGLU), and galactocerebrosidase (GALC) genes have a gene-dosage effect on Aβ40 levels in brain interstitial fluid in C57BL/6 mice and significantly increase Aβ plaque formation in the 5xFAD mouse model of AD, thus providing in vivo validation of the human genetic data. A more detailed analysis of PPT1 heterozygosity in 18-month-old mice revealed changes in α-, β-, and γ-secretases that favor an amyloidogenic pathway. Proteomic changes in brain tissue from aged PPT1 heterozygous sheep are consistent with both the mouse data and the potential activation of AD pathways. Finally, CNS-directed, AAV-mediated gene therapy significantly decreased Aβ plaques, increased life span, and improved behavioral performance in 5xFAD/PPT1+/- mice. Collectively, these data strongly suggest that heterozygosity of multiple lysosomal enzyme genes represent risk factors for AD and may identify precise therapeutic targets for a subset of genetically-defined AD patients.
Collapse
Affiliation(s)
- Bruno A Benitez
- Department of Medicine, Washington University, St. Louis, MO 63110
- Department of Psychiatry, Washington University, St. Louis, MO 63110
- Current address: Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Clare E Wallace
- Department of Neurology, Washington University, St. Louis, MO 63110
| | - Maulikkumar Patel
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | | | - Carla M Yuede
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | | | - Cyril Pottier
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | - Arda Cetin
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | - Matthew Johnson
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | - Mia T Bevan
- Department of Neurology, Washington University, St. Louis, MO 63110
| | | | - Hannah M Edwards
- Department of Neurology, Washington University, St. Louis, MO 63110
| | | | - Ryan T Harrigan
- Department of Neurology, Washington University, St. Louis, MO 63110
| | - Dominic Kurian
- Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG
| | - Thomas M Wishart
- Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG
- Current primary address: Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, School of Science and Technology, Nottingham Trent University, NHB 084, Clifton Campus, NG11 8NS
| | - Colin Smith
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - John R Cirrito
- Department of Neurology, Washington University, St. Louis, MO 63110
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110
| | - Mark S Sands
- Department of Medicine, Washington University, St. Louis, MO 63110
- Department of Genetics, Washington University, St. Louis, MO 63110
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110
| |
Collapse
|
8
|
Nixon RA, Rubinsztein DC. Mechanisms of autophagy-lysosome dysfunction in neurodegenerative diseases. Nat Rev Mol Cell Biol 2024; 25:926-946. [PMID: 39107446 DOI: 10.1038/s41580-024-00757-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/15/2024]
Abstract
Autophagy is a lysosome-based degradative process used to recycle obsolete cellular constituents and eliminate damaged organelles and aggregate-prone proteins. Their postmitotic nature and extremely polarized morphologies make neurons particularly vulnerable to disruptions caused by autophagy-lysosomal defects, especially as the brain ages. Consequently, mutations in genes regulating autophagy and lysosomal functions cause a wide range of neurodegenerative diseases. Here, we review the role of autophagy and lysosomes in neurodegenerative diseases such as Alzheimer disease, Parkinson disease and frontotemporal dementia. We also consider the strong impact of cellular ageing on lysosomes and autophagy as a tipping point for the late-age emergence of related neurodegenerative disorders. Many of these diseases have primary defects in autophagy, for example affecting autophagosome formation, and in lysosomal functions, especially pH regulation and calcium homeostasis. We have aimed to provide an integrative framework for understanding the central importance of autophagic-lysosomal function in neuronal health and disease.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| |
Collapse
|
9
|
Cissé L, Bamba S, Diallo SH, Ji W, Dembélé ME, Yalcouyé A, Coulibaly T, Traoré I, Jeffries L, Diarra S, Maiga ADB, Diallo S, Nimaga K, Touré A, Traoré O, Kotioumbé M, Mis EK, Cissé CAK, Guinto CO, Fischbeck KH, Khokha MK, Lakhani SA, Landouré G. Genetic profile of progressive myoclonic epilepsy in Mali reveals novel findings. Front Neurol 2024; 15:1455467. [PMID: 39385815 PMCID: PMC11461190 DOI: 10.3389/fneur.2024.1455467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/13/2024] [Indexed: 10/12/2024] Open
Abstract
Background and objectives Progressive myoclonic epilepsy (PME) is a group of neurological disorders characterized by recurrent myoclonic seizures with progressive neurological deterioration. We investigated the genetics of three unrelated patients with PME from Mali, a country in sub-Saharan Africa highly underrepresented in genetic and genomic research. Methods Participants were carefully examined and phenotyped. DNA was obtained for genetic analysis including whole exome sequencing (WES). In silico prediction tools and ACMG criteria were used to assess the deleteriousness of putative candidate variants. Results Pedigree analysis suggests autosomal recessive inheritance patterns for one family and sporadic forms of PME for the two other cases. WES identified novel homozygous missense variants in all the three patients, one each for NHLRC1, EPM2A, and NEU1. The sequence variants segregated with PME in each family and in silico studies including protein 3D structures, CADD scores and ACMG criteria suggested that they were damaging. Discussion PME is a group of clinically heterogeneous neurological disorders. Most reported cases in the literature are from European background with only a few cases described in North Africa. We report here novel pathogenic variants in three different genes causing PME phenotypes in three unrelated Malian patients, suggesting that genetic studies of underrepresented populations may expand the genetic epidemiology of PME. These findings also emphasize the need for inclusive genetic research to ensure a more targeted diagnostic and therapeutic approaches for diverse patient populations.
Collapse
Affiliation(s)
- Lassana Cissé
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Service de Médecine, Hôpital Nianankoro Fomba de Ségou, Ségou, Mali
| | - Salia Bamba
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Seybou H. Diallo
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Service de Neurologie, Centre Hospitalier Universitaire Gabriel Touré, Bamako, Mali
| | - Weizhen Ji
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Mohamed Emile Dembélé
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Abdoulaye Yalcouyé
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Toumany Coulibaly
- Service de Neurologie, Centre Hospitalier Universitaire Point G, Bamako, Mali
| | - Ibrahima Traoré
- Service de Neurologie, Centre Hospitalier Universitaire Gabriel Touré, Bamako, Mali
| | - Lauren Jeffries
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Salimata Diarra
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Alassane Dit Baneye Maiga
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Salimata Diallo
- Service de Neurologie, Centre Hospitalier Universitaire Gabriel Touré, Bamako, Mali
| | | | - Amadou Touré
- Service de Pédiatrie, Centre Hospitalier Universitaire Gabriel Touré, Bamako, Mali
| | - Oumou Traoré
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Mahamadou Kotioumbé
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Emily Kathryn Mis
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Cheick Abdel Kader Cissé
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Cheick Oumar Guinto
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Service de Neurologie, Centre Hospitalier Universitaire Point G, Bamako, Mali
| | | | - Mustafa K. Khokha
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Saquib A. Lakhani
- Department of Pediatrics, Pediatric Genomics Discovery Program (PGDP), Yale University School of Medicine, New Haven, CT, United States
| | - Guida Landouré
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
- Service de Neurologie, Centre Hospitalier Universitaire Point G, Bamako, Mali
| |
Collapse
|
10
|
Nixon RA. Autophagy-lysosomal-associated neuronal death in neurodegenerative disease. Acta Neuropathol 2024; 148:42. [PMID: 39259382 PMCID: PMC11418399 DOI: 10.1007/s00401-024-02799-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024]
Abstract
Autophagy, the major lysosomal pathway for degrading damaged or obsolete constituents, protects neurons by eliminating toxic organelles and peptides, restoring nutrient and energy homeostasis, and inhibiting apoptosis. These functions are especially vital in neurons, which are postmitotic and must survive for many decades while confronting mounting challenges of cell aging. Autophagy failure, especially related to the declining lysosomal ("phagy") functions, heightens the neuron's vulnerability to genetic and environmental factors underlying Alzheimer's disease (AD) and other late-age onset neurodegenerative diseases. Components of the global autophagy-lysosomal pathway and the closely integrated endolysosomal system are increasingly implicated as primary targets of these disorders. In AD, an imbalance between heightened autophagy induction and diminished lysosomal function in highly vulnerable pyramidal neuron populations yields an intracellular lysosomal build-up of undegraded substrates, including APP-βCTF, an inhibitor of lysosomal acidification, and membrane-damaging Aβ peptide. In the most compromised of these neurons, β-amyloid accumulates intraneuronally in plaque-like aggregates that become extracellular senile plaques when these neurons die, reflecting an "inside-out" origin of amyloid plaques seen in human AD brain and in mouse models of AD pathology. In this review, the author describes the importance of lysosomal-dependent neuronal cell death in AD associated with uniquely extreme autophagy pathology (PANTHOS) which is described as triggered by lysosomal membrane permeability during the earliest "intraneuronal" stage of AD. Effectors of other cell death cascades, notably calcium-activated calpains and protein kinases, contribute to lysosomal injury that induces leakage of cathepsins and activation of additional death cascades. Subsequent events in AD, such as microglial invasion and neuroinflammation, induce further cytotoxicity. In major neurodegenerative disease models, neuronal death and ensuing neuropathologies are substantially remediable by reversing underlying primary lysosomal deficits, thus implicating lysosomal failure and autophagy dysfunction as primary triggers of lysosomal-dependent cell death and AD pathogenesis and as promising therapeutic targets.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Neuroscience Institute, New York University, New York, NY, 10012, USA.
| |
Collapse
|
11
|
Du J, Shui H, Chen R, Dong Y, Xiao C, Hu Y, Wong NK. Neuraminidase-1 (NEU1): Biological Roles and Therapeutic Relevance in Human Disease. Curr Issues Mol Biol 2024; 46:8031-8052. [PMID: 39194692 DOI: 10.3390/cimb46080475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Neuraminidases catalyze the desialylation of cell-surface glycoconjugates and play crucial roles in the development and function of tissues and organs. In both physiological and pathophysiological contexts, neuraminidases mediate diverse biological activities via the catalytic hydrolysis of terminal neuraminic, or sialic acid residues in glycolipid and glycoprotein substrates. The selective modulation of neuraminidase activity constitutes a promising strategy for treating a broad spectrum of human pathologies, including sialidosis and galactosialidosis, neurodegenerative disorders, cancer, cardiovascular diseases, diabetes, and pulmonary disorders. Structurally distinct as a large family of mammalian proteins, neuraminidases (NEU1 through NEU4) possess dissimilar yet overlapping profiles of tissue expression, cellular/subcellular localization, and substrate specificity. NEU1 is well characterized for its lysosomal catabolic functions, with ubiquitous and abundant expression across such tissues as the kidney, pancreas, skeletal muscle, liver, lungs, placenta, and brain. NEU1 also exhibits a broad substrate range on the cell surface, where it plays hitherto underappreciated roles in modulating the structure and function of cellular receptors, providing a basis for it to be a potential drug target in various human diseases. This review seeks to summarize the recent progress in the research on NEU1-associated diseases and highlight the mechanistic implications of NEU1 in disease pathogenesis. An improved understanding of NEU1-associated diseases should help accelerate translational initiatives to develop novel or better therapeutics.
Collapse
Affiliation(s)
- Jingxia Du
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, China
| | - Hanqi Shui
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, China
| | - Rongjun Chen
- Clinical Pharmacology Section, Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Yibo Dong
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, China
| | - Chengyao Xiao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, China
| | - Yue Hu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471023, China
| | - Nai-Kei Wong
- Clinical Pharmacology Section, Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
12
|
van de Vlekkert D, Hu H, Weesner JA, Fremuth LE, Brown SA, Lu M, Gomero E, Campos Y, Sheppard H, d'Azzo A. AAV-mediated gene therapy for sialidosis. Mol Ther 2024; 32:2094-2112. [PMID: 38796704 PMCID: PMC11287007 DOI: 10.1016/j.ymthe.2024.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/01/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024] Open
Abstract
Sialidosis (mucolipidosis I) is a glycoprotein storage disease, clinically characterized by a spectrum of systemic and neurological phenotypes. The primary cause of the disease is deficiency of the lysosomal sialidase NEU1, resulting in accumulation of sialylated glycoproteins/oligosaccharides in tissues and body fluids. Neu1-/- mice recapitulate the severe, early-onset forms of the disease, affecting visceral organs, muscles, and the nervous system, with widespread lysosomal vacuolization evident in most cell types. Sialidosis is considered an orphan disorder with no therapy currently available. Here, we assessed the therapeutic potential of AAV-mediated gene therapy for the treatment of sialidosis. Neu1-/- mice were co-injected with two scAAV2/8 vectors, expressing human NEU1 and its chaperone PPCA. Treated mice were phenotypically indistinguishable from their WT controls. NEU1 activity was restored to different extent in most tissues, including the brain, heart, muscle, and visceral organs. This resulted in diminished/absent lysosomal vacuolization in multiple cell types and reversal of sialyl-oligosacchariduria. Lastly, normalization of lysosomal exocytosis in the cerebrospinal fluids and serum of treated mice, coupled to diminished neuroinflammation, were measures of therapeutic efficacy. These findings point to AAV-mediated gene therapy as a suitable treatment for sialidosis and possibly other diseases, associated with low NEU1 expression.
Collapse
Affiliation(s)
| | - Huimin Hu
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jason A Weesner
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leigh E Fremuth
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Scott A Brown
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Meifen Lu
- Devision of Comparative Pathology, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elida Gomero
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yvan Campos
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather Sheppard
- Devision of Comparative Pathology, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
13
|
Jang BG, Choi B, Kim MJ. Pyrogallol intermediates elicit beta-amyloid secretion via radical formation and alterations in intracellular trafficking, distinct from pyrogallol-generated superoxide. Redox Biol 2024; 73:103180. [PMID: 38795546 PMCID: PMC11140794 DOI: 10.1016/j.redox.2024.103180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/28/2024] Open
Abstract
This study unveils a novel role of pyrogallol (PG), a recognized superoxide generator, in inducing beta-amyloid (Aβ) secretion in an Alzheimer's disease (AD) cellular model. Contrary to expectations, the analysis of dihydroethidium fluorescence and UV-VIS spectrum scanning reveals that Aβ secretion arises from PG reaction intermediates rather than superoxide or other by-products. Investigation into Aβ secretion mechanisms identifies dynasore-dependent endocytosis and BFA-dependent exocytosis as independent pathways, regulated by tiron, tempol, and superoxide dismutase. Cell-type specificity is observed, with 293sw cells showing both pathways, while H4sw cells and primary astrocytes from an AD animal model exclusively exhibit the Aβ exocytosis pathway. This exploration contributes to understanding PG's chemical reactions and provides insights into the interplay between environmental factors, free radicals, and AD, linking occupational PG exposure to AD risk as reported in the literature.
Collapse
Affiliation(s)
- Bong-Geum Jang
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Boyoung Choi
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Min-Ju Kim
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea; Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
14
|
Glynn C, Rodriguez JA, Hyman BT. The structural line between prion and "prion-like": Insights from prion protein and tau. Curr Opin Neurobiol 2024; 86:102857. [PMID: 38489865 PMCID: PMC11162956 DOI: 10.1016/j.conb.2024.102857] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/17/2024]
Abstract
The concept of 'prion-like' behavior has emerged in the study of diseases involving protein misfolding where fibrillar structures, called amyloids, self-propagate and induce disease in a fashion similar to prions. From a biological standpoint, in order to be considered 'prion-like,' a protein must traverse cells and tissues and further propagate via a templated conformational change. Since 2017, cryo-electron microscopy structures from patient-derived 'prion-like' amyloids, in particular tau, have been presented and revealed structural similarities shared across amyloids. Since 2021, cryo-EM structures from prions of known infectivity have been added to the ex vivo amyloid structure family. In this review, we discuss current proposals for the 'prion-like' mechanisms of spread for tau and prion protein as well as discuss different influencers on structures of aggregates from tauopathies and prion diseases. Lastly, we discuss some of the current hypotheses for what may distinguish structures that are 'prion-like' from transmissible prion structures.
Collapse
Affiliation(s)
- Calina Glynn
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Cambridge, MA, USA
| | - Jose A Rodriguez
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics, STROBE, NSF Science and Technology Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
15
|
Fremuth LE, Hu H, van de Vlekkert D, Annunziata I, Weesner JA, Gomero E, d'Azzo A. Neuraminidase 1 regulates the cellular state of microglia by modulating the sialylation of Trem2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595036. [PMID: 38826426 PMCID: PMC11142087 DOI: 10.1101/2024.05.20.595036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Neuraminidase 1 (Neu1) cleaves terminal sialic acids from sialoglycoproteins in endolysosomes and at the plasma membrane. As such, Neu1 regulates immune cells, primarily those of the monocytic lineage. Here we examined how Neu1 influences microglia by modulating the sialylation of full-length Trem2 (Trem2-FL), a multifunctional receptor that regulates microglial survival, phagocytosis, and cytokine production. When Neu1 was deficient/downregulated, Trem2-FL remained sialylated, accumulated intracellularly, and was excessively cleaved into a C-terminal fragment (Trem2-CTF) and an extracellular soluble domain (sTrem2), enhancing their signaling capacities. Sialylated Trem2-FL (Sia-Trem2-FL) did not hinder Trem2-FL-DAP12-Syk complex assembly but impaired signal transduction through Syk, ultimately abolishing Trem2-dependent phagocytosis. Concurrently, Trem2-CTF-DAP12 complexes dampened NFκB signaling, while sTrem2 propagated Akt-dependent cell survival and NFAT1-mediated production of TNFα and CCL3. Because Neu1 and Trem2 are implicated in neurodegenerative/neuroinflammatory diseases, including Alzheimer disease (AD) and sialidosis, modulating Neu1 activity represents a therapeutic approach to broadly regulate microglia-mediated neuroinflammation.
Collapse
|
16
|
Xu T, Heon-Roberts R, Moore T, Dubot P, Pan X, Guo T, Cairo CW, Holley R, Bigger B, Durcan TM, Levade T, Ausseil J, Amilhon B, Gorelik A, Nagar B, Sturiale L, Palmigiano A, Röckle I, Thiesler H, Hildebrandt H, Garozzo D, Pshezhetsky AV. Secondary deficiency of neuraminidase 1 contributes to CNS pathology in neurological mucopolysaccharidoses via hypersialylation of brain glycoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.587986. [PMID: 38712143 PMCID: PMC11071461 DOI: 10.1101/2024.04.26.587986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Mucopolysaccharidoses (MPS) are lysosomal storage diseases caused by defects in catabolism of glycosaminoglycans. MPS I, II, III and VII are associated with lysosomal accumulation of heparan sulphate and manifest with neurological deterioration. Most of these neurological MPS currently lack effective treatments. Here, we report that, compared to controls, neuraminidase 1 (NEU1) activity is drastically reduced in brain tissues of neurological MPS patients and in mouse models of MPS I, II, IIIA, IIIB and IIIC, but not of other neurological lysosomal disorders not presenting with heparan sulphate storage. We further show that accumulated heparan sulphate disrupts the lysosomal multienzyme complex of NEU1 with cathepsin A (CTSA), β-galactosidase (GLB1) and glucosamine-6-sulfate sulfatase (GALNS) necessary to maintain enzyme activity, and that NEU1 deficiency is linked to partial deficiencies of GLB1 and GALNS in cortical tissues and iPSC-derived cortical neurons of neurological MPS patients. Increased sialylation of N-linked glycans in brain samples of human MPS III patients and MPS IIIC mice implicated insufficient processing of brain N-linked sialylated glycans, except for polysialic acid, which was reduced in the brains of MPS IIIC mice. Correction of NEU1 activity in MPS IIIC mice by lentiviral gene transfer ameliorated previously identified hallmarks of the disease, including memory impairment, behavioural traits, and reduced levels of the excitatory synapse markers VGLUT1 and PSD95. Overexpression of NEU1 also restored levels of VGLUT1-/PSD95-positive puncta in cortical neurons derived from iPSC of an MPS IIIA patient. Together, our data demonstrate that heparan sulphate-induced secondary NEU1 deficiency and aberrant sialylation of glycoproteins implicated in synaptogenesis, memory, and behaviour constitute a novel pathological pathway in neurological MPS spectrum crucially contributing to CNS pathology. Graphical abstract
Collapse
|
17
|
Inci OK, Basırlı H, Can M, Yanbul S, Seyrantepe V. Gangliosides as Therapeutic Targets for Neurodegenerative Diseases. J Lipids 2024; 2024:4530255. [PMID: 38623278 PMCID: PMC11018381 DOI: 10.1155/2024/4530255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/05/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
Gangliosides, sialic acid-containing glycosphingolipids, are abundant in cell membranes and primarily involved in controlling cell signaling and cell communication. The altered ganglioside pattern has been demonstrated in several neurodegenerative diseases, characterized during early-onset or infancy, emphasizing the significance of gangliosides in the brain. Enzymes required for the biosynthesis of gangliosides are linked to several devastating neurological disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), hereditary spastic paraplegia (HSP). In this review, we summarized not only the critical roles of biosynthetic enzymes and their inhibitors in ganglioside metabolism but also the efficacy of treatment strategies of ganglioside to address their significance in those diseases.
Collapse
Affiliation(s)
- Orhan Kerim Inci
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce Campus, Urla, 35430 Izmir, Türkiye
| | - Hande Basırlı
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce Campus, Urla, 35430 Izmir, Türkiye
| | - Melike Can
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce Campus, Urla, 35430 Izmir, Türkiye
| | - Selman Yanbul
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce Campus, Urla, 35430 Izmir, Türkiye
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce Campus, Urla, 35430 Izmir, Türkiye
- Izmir Institute of Technology, IYTEDEHAM, Gulbahce Campus, Urla, 35430 Izmir, Türkiye
| |
Collapse
|
18
|
Yang X, Jiao Y, Zhang Y, Sun M, Gao Y, Zhou Y, Xiao H, Ren J, Zhou Z, Zhai Y, Song B, Zhang L, Kong P. Oseltamivir enhances 5-FU sensitivity in esophageal squamous carcinoma with high SPNS1. Biomed Pharmacother 2024; 173:116367. [PMID: 38460365 DOI: 10.1016/j.biopha.2024.116367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
Sphingolipid transporter 1 (SPNS1) is a significant differentially expressed gene (DEGs) in esophageal squamous cell carcinoma (ESCC). According to 3 pairs clinic cohorts, transcriptomic (155 pairs of ESCC samples and GSE53624, and proteomic data from PXD021701 including 124 ESCC samples) we found that SPNS1 was significantly higher in ESCC tissues compared to adjacent normal esophagus tissues. ESCC patients with high SPNS1 had a significantly poorer clinical prognosis than those with low SPNS1. Knockdown of SPNS1 significantly inhibited the proliferation, migration, and invasion abilities of ESCC cells, while promoting apoptosis. And overexpression of SPNS1 exhibited opposite functions. Furthermore, ESCC cells became more sensitive to 5-fluorouracil (5-FU) when SPNS1 was knocked down. Transcriptome sequencing revealed that NEU1 was one significant DEG affected by SPNS1 and positively correlated with SPNS1 expression. Oseltamivir phosphate (OP), one NEU1 inhibitor, markedly reversed 5-FU resistance, migration, and proliferation induced by high expression of SPNS1 both in vivo and in vitro. Our findings indicated that SPNS1 might promote the progression of ESCC by upregulating NEU1 expression and influencing chemotherapy sensitivity. These results provide new perceptions into potential therapeutic targets for ESCC treatment. The present study aimed to investigate the role and underlying mechanism of SPNS1 in ESCC.
Collapse
Affiliation(s)
- Xin Yang
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ye Jiao
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingying Zhang
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meng Sun
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingzhen Gao
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Zhou
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Heng Xiao
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Ren
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhinan Zhou
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuanfang Zhai
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Human Anatomy, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Bin Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi, China
| | - Ling Zhang
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Pengzhou Kong
- Translational Medicine Research Center, Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research of Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, China; Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China; State Key Laboratory for Pneumoconiosis of National Health Commission, Key Laboratory of Prevention, Treatment and Fundamental Studies for Respiratory Diseases of Shanxi, Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
19
|
Wißfeld J, Abou Assale T, Cuevas-Rios G, Liao H, Neumann H. Therapeutic potential to target sialylation and SIGLECs in neurodegenerative and psychiatric diseases. Front Neurol 2024; 15:1330874. [PMID: 38529039 PMCID: PMC10961342 DOI: 10.3389/fneur.2024.1330874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
Sialic acids, commonly found as the terminal carbohydrate on the glycocalyx of mammalian cells, are pivotal checkpoint inhibitors of the innate immune system, particularly within the central nervous system (CNS). Sialic acid-binding immunoglobulin-like lectins (SIGLECs) expressed on microglia are key players in maintaining microglial homeostasis by recognizing intact sialylation. The finely balanced sialic acid-SIGLEC system ensures the prevention of excessive and detrimental immune responses in the CNS. However, loss of sialylation and SIGLEC receptor dysfunctions contribute to several chronic CNS diseases. Genetic variants of SIGLEC3/CD33, SIGLEC11, and SIGLEC14 have been associated with neurodegenerative diseases such as Alzheimer's disease, while sialyltransferase ST8SIA2 and SIGLEC4/MAG have been linked to psychiatric diseases such as schizophrenia, bipolar disorders, and autism spectrum disorders. Consequently, immune-modulatory functions of polysialic acids and SIGLEC binding antibodies have been exploited experimentally in animal models of Alzheimer's disease and inflammation-induced CNS tissue damage, including retinal damage. While the potential of these therapeutic approaches is evident, only a few therapies to target either sialylation or SIGLEC receptors have been tested in patient clinical trials. Here, we provide an overview of the critical role played by the sialic acid-SIGLEC axis in shaping microglial activation and function within the context of neurodegeneration and synaptopathies and discuss the current landscape of therapies that target sialylation or SIGLECs.
Collapse
Affiliation(s)
- Jannis Wißfeld
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Tawfik Abou Assale
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - German Cuevas-Rios
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Huan Liao
- Florey Institute of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
20
|
Settembre C, Perera RM. Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat Rev Mol Cell Biol 2024; 25:223-245. [PMID: 38001393 DOI: 10.1038/s41580-023-00676-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/26/2023]
Abstract
Every cell must satisfy basic requirements for nutrient sensing, utilization and recycling through macromolecular breakdown to coordinate programmes for growth, repair and stress adaptation. The lysosome orchestrates these key functions through the synchronised interplay between hydrolytic enzymes, nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles and regulation of specific gene expression programmes. In this Review, we discuss recent findings on lysosome-dependent signalling pathways, focusing on how the lysosome senses nutrient availability through its physical and functional association with mechanistic target of rapamycin complex 1 (mTORC1) and how, in response, the microphthalmia/transcription factor E (MiT/TFE) transcription factors exert feedback regulation on lysosome biogenesis. We also highlight the emerging interactions of lysosomes with other organelles, which contribute to cellular homeostasis. Lastly, we discuss how lysosome dysfunction contributes to diverse disease pathologies and how inherited mutations that compromise lysosomal hydrolysis, transport or signalling components lead to multi-organ disorders with severe metabolic and neurological impact. A deeper comprehension of lysosomal composition and function, at both the cellular and organismal level, may uncover fundamental insights into human physiology and disease.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Rushika M Perera
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Jeong J, Kim OH, Shim J, Keum S, Hwang YE, Song S, Kim JW, Choi JH, Lee HJ, Rhee S. Microtubule acetylation induced by oxidative stress regulates subcellular distribution of lysosomal vesicles for amyloid-beta secretion. J Cell Physiol 2023; 238:2812-2826. [PMID: 37801327 DOI: 10.1002/jcp.31131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Abstract
Excessive production and accumulation of amyloid-beta (Aβ) in the brain are one of the hallmarks of Alzheimer's disease (AD). Although oxidative stress is known to trigger and promote the progression of AD, the molecular relationship between oxidative stress and Aβ production is not yet fully understood. In this study, we demonstrate that microtubule acetylation induced by oxidative stress plays a critical role in Aβ production and secretion by altering the subcellular distribution of Aβ precursor protein (APP)-containing lysosomal vesicles. Under oxidative stress, both H4-APPSwe/Ind and HEK293T-APPSwe/Ind cell lines showed increased microtubule acetylation and Aβ secretion. Knockdown (KD) of alpha-tubulin N-acetyltransferase 1 (ATAT1) by using a lentiviral shRNA not only inhibited the generation of intermediate APP fragments, such as β-CTF and AICD, but also suppressed Aβ secretion. Oxidative stress promoted the dispersion of LAMP1-positive vesicles to the periphery of the cell through microtubule acetylation, leading to the formation of neutralized lysosomal vesicles (NLVs), which was inhibited by ATAT1 KD. Treatment of the cells with the dynein ATPase inhibitor EHNA or downregulation of LIS1, a regulator of dynein-mediated intracellular transport, increased the peripheral localization of NLVs and promoted Aβ secretion, whereas KD of ADP ribosylation factor like GTPase 8B showed the opposite result. ATAT1 KD in the hippocampal region of the 5×FAD AD mouse model also showed significant reductions in Aβ plaque accumulation and memory loss. Taken together, these findings suggest that oxidative stress-induced microtubule acetylation promotes the peripheral localization of lysosomal vesicles to form NLVs, thereby enhancing Aβ secretion.
Collapse
Affiliation(s)
- Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jaeyeoung Shim
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Seula Keum
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Ye Eun Hwang
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Seongeun Song
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Jee-Hye Choi
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Itoh K, Tsukimoto J. Lysosomal sialidase NEU1, its intracellular properties, deficiency, and use as a therapeutic agent. Glycoconj J 2023; 40:611-619. [PMID: 38147151 DOI: 10.1007/s10719-023-10135-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 12/27/2023]
Abstract
Neuraminidase 1 (NEU1) is a lysosomal sialidase that cleaves terminal α-linked sialic acid residues from sialylglycans. NEU1 is biosynthesized in the rough endoplasmic reticulum (RER) lumen as an N-glycosylated protein to associate with its protective protein/cathepsin A (CTSA) and then form a lysosomal multienzyme complex (LMC) also containing β-galactosidase 1 (GLB1). Unlike other mammalian sialidases, including NEU2 to NEU4, NEU1 transport to lysosomes requires association of NEU1 with CTSA, binding of the CTSA carrying terminal mannose 6-phosphate (M6P)-type N-glycan with M6P receptor (M6PR), and intralysosomal NEU1 activation at acidic pH. In contrast, overexpression of the single NEU1 gene in mammalian cells causes intracellular NEU1 protein crystallization in the RER due to self-aggregation when intracellular CTSA is reduced to a relatively low level. Sialidosis (SiD) and galactosialidosis (GS) are autosomal recessive lysosomal storage diseases caused by the gene mutations of NEU1 and CTSA, respectively. These incurable diseases associate with the NEU1 deficiency, excessive accumulation of sialylglycans in neurovisceral organs, and systemic manifestations. We established a novel GS model mouse carrying homozygotic Ctsa IVS6 + 1 g/a mutation causing partial exon 6 skipping with simultaneous deficiency of Ctsa and Neu1. Symptoms developed in the GS mice like those in juvenile/adult GS patients, such as myoclonic seizures, suppressed behavior, gargoyle-like face, edema, proctoptosis due to Neu1 deficiency, and sialylglycan accumulation associated with neurovisceral inflammation. We developed a modified NEU1 (modNEU1), which does not form protein crystals but is transported to lysosomes by co-expressed CTSA. In vivo gene therapy for GS and SiD utilizing a single adeno-associated virus (AAV) carrying modNEU1 and CTSA genes under dual promoter control will be created.
Collapse
Affiliation(s)
- Kohji Itoh
- Department of Medicinal Biotechnology, Institute for Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan.
| | - Jun Tsukimoto
- Department of Medicinal Biotechnology, Institute for Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
23
|
van de Vlekkert D, Hu H, Fremuth LE, Brown SA, Weesner JA, Gomero E, Campos Y, d'Azzo A. AAV-mediated gene therapy for Sialidosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566667. [PMID: 38014061 PMCID: PMC10680618 DOI: 10.1101/2023.11.10.566667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Sialidosis is a glycoprotein storage disease caused by deficiency of the lysosomal sialidase NEU1, which leads to pathogenic accumulation of sialylated glycoproteins and oligosaccharides in tissues and body fluids. The disease belongs to the group of orphan disorders with no therapy currently available. Here, we have tested the therapeutic potential of AAV-mediated gene therapy for the treatment of sialidosis in a mouse model of the disease. One-month-old Neu1 -/- mice were co-injected with two scAAV2/8 vectors, expressing NEU1 and its chaperone PPCA, and sacrificed at 3 months post-injection. Treated mice were phenotypically indistinguishable from their WT controls. Histopathologically, they showed diminished or absent vacuolization in cells of visceral organs, including the kidney, as well as the choroid plexus and other areas of the brain. This was accompanied by restoration of NEU1 activity in most tissues, reversal of sialyl-oligosacchariduria, and normalization of lysosomal exocytosis in the CSF and serum of treated mice. AAV injection prevented the occurrence of generalized fibrosis, which is a prominent contributor of disease pathogenesis in Neu1 -/- mice and likely in patients. Overall, this therapeutic strategy holds promise for the treatment of sialidosis and may be applicable to adult forms of human idiopathic fibrosis with low NEU1 expression.
Collapse
|
24
|
Jiang Y, MacNeil LT. Simple model systems reveal conserved mechanisms of Alzheimer's disease and related tauopathies. Mol Neurodegener 2023; 18:82. [PMID: 37950311 PMCID: PMC10638731 DOI: 10.1186/s13024-023-00664-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023] Open
Abstract
The lack of effective therapies that slow the progression of Alzheimer's disease (AD) and related tauopathies highlights the need for a more comprehensive understanding of the fundamental cellular mechanisms underlying these diseases. Model organisms, including yeast, worms, and flies, provide simple systems with which to investigate the mechanisms of disease. The evolutionary conservation of cellular pathways regulating proteostasis and stress response in these organisms facilitates the study of genetic factors that contribute to, or protect against, neurodegeneration. Here, we review genetic modifiers of neurodegeneration and related cellular pathways identified in the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, and the fruit fly Drosophila melanogaster, focusing on models of AD and related tauopathies. We further address the potential of simple model systems to better understand the fundamental mechanisms that lead to AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuwei Jiang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
25
|
Boeddrich A, Haenig C, Neuendorf N, Blanc E, Ivanov A, Kirchner M, Schleumann P, Bayraktaroğlu I, Richter M, Molenda CM, Sporbert A, Zenkner M, Schnoegl S, Suenkel C, Schneider LS, Rybak-Wolf A, Kochnowsky B, Byrne LM, Wild EJ, Nielsen JE, Dittmar G, Peters O, Beule D, Wanker EE. A proteomics analysis of 5xFAD mouse brain regions reveals the lysosome-associated protein Arl8b as a candidate biomarker for Alzheimer's disease. Genome Med 2023; 15:50. [PMID: 37468900 PMCID: PMC10357615 DOI: 10.1186/s13073-023-01206-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/22/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by the intra- and extracellular accumulation of amyloid-β (Aβ) peptides. How Aβ aggregates perturb the proteome in brains of patients and AD transgenic mouse models, remains largely unclear. State-of-the-art mass spectrometry (MS) methods can comprehensively detect proteomic alterations, providing relevant insights unobtainable with transcriptomics investigations. Analyses of the relationship between progressive Aβ aggregation and protein abundance changes in brains of 5xFAD transgenic mice have not been reported previously. METHODS We quantified progressive Aβ aggregation in hippocampus and cortex of 5xFAD mice and controls with immunohistochemistry and membrane filter assays. Protein changes in different mouse tissues were analyzed by MS-based proteomics using label-free quantification; resulting MS data were processed using an established pipeline. Results were contrasted with existing proteomic data sets from postmortem AD patient brains. Finally, abundance changes in the candidate marker Arl8b were validated in cerebrospinal fluid (CSF) from AD patients and controls using ELISAs. RESULTS Experiments revealed faster accumulation of Aβ42 peptides in hippocampus than in cortex of 5xFAD mice, with more protein abundance changes in hippocampus, indicating that Aβ42 aggregate deposition is associated with brain region-specific proteome perturbations. Generating time-resolved data sets, we defined Aβ aggregate-correlated and anticorrelated proteome changes, a fraction of which was conserved in postmortem AD patient brain tissue, suggesting that proteome changes in 5xFAD mice mimic disease-relevant changes in human AD. We detected a positive correlation between Aβ42 aggregate deposition in the hippocampus of 5xFAD mice and the abundance of the lysosome-associated small GTPase Arl8b, which accumulated together with axonal lysosomal membranes in close proximity of extracellular Aβ plaques in 5xFAD brains. Abnormal aggregation of Arl8b was observed in human AD brain tissue. Arl8b protein levels were significantly increased in CSF of AD patients. CONCLUSIONS We report a comprehensive biochemical and proteomic investigation of hippocampal and cortical brain tissue derived from 5xFAD transgenic mice, providing a valuable resource to the neuroscientific community. We identified Arl8b, with significant abundance changes in 5xFAD and AD patient brains. Arl8b might enable the measurement of progressive lysosome accumulation in AD patients and have clinical utility as a candidate biomarker.
Collapse
Affiliation(s)
- Annett Boeddrich
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Christian Haenig
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Nancy Neuendorf
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Andranik Ivanov
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité - University Medicine Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Philipp Schleumann
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Irem Bayraktaroğlu
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Matthias Richter
- Advanced Light Microscopy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Christine Mirjam Molenda
- Advanced Light Microscopy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Martina Zenkner
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Sigrid Schnoegl
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Christin Suenkel
- Systems Biology of Gene Regulatory Elements, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Luisa-Sophie Schneider
- Department of Psychiatry, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Agnieszka Rybak-Wolf
- Systems Biology of Gene Regulatory Elements, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Bianca Kochnowsky
- Department of Psychiatry, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Lauren M Byrne
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Edward J Wild
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- National Hospital for Neurology & Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Jørgen E Nielsen
- Neurogenetics Clinic & Research Lab, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Section 8008, Inge Lehmanns Vej 8, 2100, Copenhagen, Denmark
| | - Gunnar Dittmar
- Core Unit Proteomics, Berlin Institute of Health at Charité - University Medicine Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Proteomics of Cellular Signalling, Luxembourg Institute of Health, 1a Rue Thomas Edison, 1445, Strassen, Luxembourg
| | - Oliver Peters
- Department of Psychiatry, Charité - University Medicine Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, 10117, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Erich E Wanker
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany.
| |
Collapse
|
26
|
Fastenau C, Wickline JL, Smith S, Odfalk KF, Solano L, Bieniek KF, Hopp SC. Increased α-2,6 sialic acid on microglia in amyloid pathology is resistant to oseltamivir. GeroScience 2023; 45:1539-1555. [PMID: 36867284 PMCID: PMC10400525 DOI: 10.1007/s11357-023-00761-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Terminal sialic acid residues are present on most glycoproteins and glycolipids, but levels of sialylation are known to change in the brain throughout the lifespan as well as during disease. Sialic acids are important for numerous cellular processes including cell adhesion, neurodevelopment, and immune regulation as well as pathogen invasion into host cells. Neuraminidase enzymes, also known as sialidases, are responsible for removal of terminal sialic acids in a process known as desialylation. Neuraminidase 1 (Neu1) cleaves the α-2,6 bond of terminal sialic acids. Aging individuals with dementia are often treated with the antiviral medication oseltamivir, which is associated with induction of adverse neuropsychiatric side effects; this drug inhibits both viral and mammalian Neu1. The present study tested whether a clinically relevant antiviral dosing regimen of oseltamivir would disrupt behavior in the 5XFAD mouse model of Alzheimer's disease amyloid pathology or wild-type littermates. While oseltamivir treatment did not impact mouse behavior or modify amyloid plaque size or morphology, a novel spatial distribution of α-2,6 sialic acid residues was discovered in 5XFAD mice that was not present in wild-type littermates. Further analyses revealed that α-2,6 sialic acid residues were not localized the amyloid plaques but instead localized to plaque-associated microglia. Notably, treatment with oseltamivir did not alter α-2,6 sialic acid distribution on plaque-associated microglia in 5XFAD mice which may be due to downregulation of Neu1 transcript levels in 5XFAD mice. Overall, this study suggests that plaque-associated microglia are highly sialylated and are resistant to change with oseltamivir, thus interfering with microglia immune recognition of and response to amyloid pathology.
Collapse
Affiliation(s)
- Caitlyn Fastenau
- Department of Pharmacology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Jessica L Wickline
- Department of Pharmacology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sabrina Smith
- Department of Pharmacology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kristian F Odfalk
- Department of Pharmacology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Leigh Solano
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
27
|
Gorelik A, Illes K, Mazhab-Jafari MT, Nagar B. Structure of the immunoregulatory sialidase NEU1. SCIENCE ADVANCES 2023; 9:eadf8169. [PMID: 37205763 DOI: 10.1126/sciadv.adf8169] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Sialic acids linked to glycoproteins and glycolipids are important mediators of cell and protein recognition events. These sugar residues are removed by neuraminidases (sialidases). Neuraminidase-1 (sialidase-1 or NEU1) is a ubiquitously expressed mammalian sialidase located in lysosomes and on the cell membrane. Because of its modulation of multiple signaling processes, it is a potential therapeutic target for cancers and immune disorders. Genetic defects in NEU1 or in its protective protein cathepsin A (PPCA, CTSA) cause the lysosomal storage diseases sialidosis and galactosialidosis. To further our understanding of this enzyme's function at the molecular level, we determined the three-dimensional structure of murine NEU1. The enzyme oligomerizes through two self-association interfaces and displays a wide substrate-binding cavity. A catalytic loop adopts an inactive conformation. We propose a mechanism of activation involving a conformational change in this loop upon binding to its protective protein. These findings may facilitate the development of selective inhibitor and agonist therapies.
Collapse
Affiliation(s)
- Alexei Gorelik
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Katalin Illes
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Mohammad T Mazhab-Jafari
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Bhushan Nagar
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Muraleedharan A, Vanderperre B. The endo-lysosomal system in Parkinson's disease: expanding the horizon. J Mol Biol 2023:168140. [PMID: 37148997 DOI: 10.1016/j.jmb.2023.168140] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence is increasing with age. A wealth of genetic evidence indicates that the endo-lysosomal system is a major pathway driving PD pathogenesis with a growing number of genes encoding endo-lysosomal proteins identified as risk factors for PD, making it a promising target for therapeutic intervention. However, detailed knowledge and understanding of the molecular mechanisms linking these genes to the disease are available for only a handful of them (e.g. LRRK2, GBA1, VPS35). Taking on the challenge of studying poorly characterized genes and proteins can be daunting, due to the limited availability of tools and knowledge from previous literature. This review aims at providing a valuable source of molecular and cellular insights into the biology of lesser-studied PD-linked endo-lysosomal genes, to help and encourage researchers in filling the knowledge gap around these less popular genetic players. Specific endo-lysosomal pathways discussed range from endocytosis, sorting, and vesicular trafficking to the regulation of membrane lipids of these membrane-bound organelles and the specific enzymatic activities they contain. We also provide perspectives on future challenges that the community needs to tackle and propose approaches to move forward in our understanding of these poorly studied endo-lysosomal genes. This will help harness their potential in designing innovative and efficient treatments to ultimately re-establish neuronal homeostasis in PD but also other diseases involving endo-lysosomal dysfunction.
Collapse
Affiliation(s)
- Amitha Muraleedharan
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| | - Benoît Vanderperre
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| |
Collapse
|
29
|
Weng IC, Chen HL, Lin WH, Liu FT. Sialylation of cell surface glycoconjugates modulates cytosolic galectin-mediated responses upon organelle damage : Minireview. Glycoconj J 2023; 40:295-303. [PMID: 37052731 DOI: 10.1007/s10719-023-10112-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/26/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
Sialylation is an important terminal modification of glycoconjugates that mediate diverse functions in physiology and disease. In this review we focus on how altered cell surface sialylation status is sensed by cytosolic galectins when the integrity of intracellular vesicles or organelles is compromised to expose luminal glycans to the cytosolic milieu, and how this impacts galectin-mediated cellular responses. In addition, we discuss the roles of mammalian sialidases on the cell surface, in the organelle lumen and cytosol, and raise the possibility that intracellular glycan processing may be critical in controlling various galectin-mediated responses when cells encounter stress.
Collapse
Affiliation(s)
- I-Chun Weng
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung-Lin Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wei-Han Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
30
|
Chen QQ, Liu K, Shi N, Ma G, Wang P, Xie HM, Jin SJ, Wei TT, Yu XY, Wang Y, Zhang JY, Li P, Qi LW, Zhang L. Neuraminidase 1 promotes renal fibrosis development in male mice. Nat Commun 2023; 14:1713. [PMID: 36973294 PMCID: PMC10043283 DOI: 10.1038/s41467-023-37450-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
The functions of the influenza virus neuraminidase has been well documented but those of the mammalian neuraminidases remain less explored. Here, we characterize the role of neuraminidase 1 (NEU1) in unilateral ureteral obstruction (UUO) and folic acid (FA)-induced renal fibrosis mouse models. We find that NEU1 is significantly upregulated in the fibrotic kidneys of patients and mice. Functionally, tubular epithelial cell-specific NEU1 knockout inhibits epithelial-to-mesenchymal transition, inflammatory cytokines production, and collagen deposition in mice. Conversely, NEU1 overexpression exacerbates progressive renal fibrosis. Mechanistically, NEU1 interacts with TGFβ type I receptor ALK5 at the 160-200aa region and stabilizes ALK5 leading to SMAD2/3 activation. Salvianolic acid B, a component of Salvia miltiorrhiza, is found to strongly bind to NEU1 and effectively protect mice from renal fibrosis in a NEU1-dependent manner. Collectively, this study characterizes a promotor role for NEU1 in renal fibrosis and suggests a potential avenue of targeting NEU1 to treat kidney diseases.
Collapse
Affiliation(s)
- Qian-Qian Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kang Liu
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ning Shi
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Gaoxiang Ma
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China
| | - Peipei Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Hua-Mei Xie
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China
| | - Si-Jia Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ting-Ting Wei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiang-Yu Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi Wang
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China
| | - Jun-Yuan Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Lian-Wen Qi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China.
| | - Lei Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
31
|
Sandhoff R, Sandhoff K. Neuronal Ganglioside and Glycosphingolipid (GSL) Metabolism and Disease : Cascades of Secondary Metabolic Errors Can Generate Complex Pathologies (in LSDs). ADVANCES IN NEUROBIOLOGY 2023; 29:333-390. [PMID: 36255681 DOI: 10.1007/978-3-031-12390-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glycosphingolipids (GSLs) are a diverse group of membrane components occurring mainly on the surfaces of mammalian cells. They and their metabolites have a role in intercellular communication, serving as versatile biochemical signals (Kaltner et al, Biochem J 476(18):2623-2655, 2019) and in many cellular pathways. Anionic GSLs, the sialic acid containing gangliosides (GGs), are essential constituents of neuronal cell surfaces, whereas anionic sulfatides are key components of myelin and myelin forming oligodendrocytes. The stepwise biosynthetic pathways of GSLs occur at and lead along the membranes of organellar surfaces of the secretory pathway. After formation of the hydrophobic ceramide membrane anchor of GSLs at the ER, membrane-spanning glycosyltransferases (GTs) of the Golgi and Trans-Golgi network generate cell type-specific GSL patterns for cellular surfaces. GSLs of the cellular plasma membrane can reach intra-lysosomal, i.e. luminal, vesicles (ILVs) by endocytic pathways for degradation. Soluble glycoproteins, the glycosidases, lipid binding and transfer proteins and acid ceramidase are needed for the lysosomal catabolism of GSLs at ILV-membrane surfaces. Inherited mutations triggering a functional loss of glycosylated lysosomal hydrolases and lipid binding proteins involved in GSL degradation cause a primary lysosomal accumulation of their non-degradable GSL substrates in lysosomal storage diseases (LSDs). Lipid binding proteins, the SAPs, and the various lipids of the ILV-membranes regulate GSL catabolism, but also primary storage compounds such as sphingomyelin (SM), cholesterol (Chol.), or chondroitin sulfate can effectively inhibit catabolic lysosomal pathways of GSLs. This causes cascades of metabolic errors, accumulating secondary lysosomal GSL- and GG- storage that can trigger a complex pathology (Breiden and Sandhoff, Int J Mol Sci 21(7):2566, 2020).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Konrad Sandhoff
- LIMES, c/o Kekule-Institute for Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany.
| |
Collapse
|
32
|
Mächtel R, Boros FA, Dobert JP, Arnold P, Zunke F. From Lysosomal Storage Disorders to Parkinson's Disease - Challenges and Opportunities. J Mol Biol 2022:167932. [PMID: 36572237 DOI: 10.1016/j.jmb.2022.167932] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Lysosomes are specialized organelles with an acidic pH that act as recycling hubs for intracellular and extracellular components. They harbour numerous different hydrolytic enzymes to degrade substrates like proteins, peptides, and glycolipids. Reduced catalytic activity of lysosomal enzymes can cause the accumulation of these substrates and loss of lysosomal integrity, resulting in lysosomal dysfunction and lysosomal storage disorders (LSDs). Post-mitotic cells, such as neurons, seem to be highly sensitive to damages induced by lysosomal dysfunction, thus LSDs often manifest with neurological symptoms. Interestingly, some LSDs and Parkinson's disease (PD) share common cellular pathomechanisms, suggesting convergence of aetiology of the two disease types. This is further underlined by genetic associations of several lysosomal genes involved in LSDs with PD. The increasing number of lysosome-associated genetic risk factors for PD makes it necessary to understand functions and interactions of lysosomal proteins/enzymes both in health and disease, thereby holding the potential to identify new therapeutic targets. In this review, we highlight genetic and mechanistic interactions between the complex lysosomal network, LSDs and PD, and elaborate on methodical challenges in lysosomal research.
Collapse
Affiliation(s)
- Rebecca Mächtel
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | | | - Jan Philipp Dobert
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Friederike Zunke
- Department of Molecular Neurology, University Clinics Erlangen, Erlangen, Germany.
| |
Collapse
|
33
|
Scerra G, De Pasquale V, Scarcella M, Caporaso MG, Pavone LM, D'Agostino M. Lysosomal positioning diseases: beyond substrate storage. Open Biol 2022; 12:220155. [PMID: 36285443 PMCID: PMC9597170 DOI: 10.1098/rsob.220155] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Lysosomal storage diseases (LSDs) comprise a group of inherited monogenic disorders characterized by lysosomal dysfunctions due to undegraded substrate accumulation. They are caused by a deficiency in specific lysosomal hydrolases involved in cellular catabolism, or non-enzymatic proteins essential for normal lysosomal functions. In LSDs, the lack of degradation of the accumulated substrate and its lysosomal storage impairs lysosome functions resulting in the perturbation of cellular homeostasis and, in turn, the damage of multiple organ systems. A substantial number of studies on the pathogenesis of LSDs has highlighted how the accumulation of lysosomal substrates is only the first event of a cascade of processes including the accumulation of secondary metabolites and the impairment of cellular trafficking, cell signalling, autophagic flux, mitochondria functionality and calcium homeostasis, that significantly contribute to the onset and progression of these diseases. Emerging studies on lysosomal biology have described the fundamental roles of these organelles in a variety of physiological functions and pathological conditions beyond their canonical activity in cellular waste clearance. Here, we discuss recent advances in the knowledge of cellular and molecular mechanisms linking lysosomal positioning and trafficking to LSDs.
Collapse
Affiliation(s)
- Gianluca Scerra
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Valeria De Pasquale
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy
| | - Melania Scarcella
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Maria Gabriella Caporaso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Massimo D'Agostino
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|
34
|
Heimerl M, Gausepohl T, Mueller JH, Ricke-Hoch M. Neuraminidases-Key Players in the Inflammatory Response after Pathophysiological Cardiac Stress and Potential New Therapeutic Targets in Cardiac Disease. BIOLOGY 2022; 11:biology11081229. [PMID: 36009856 PMCID: PMC9405403 DOI: 10.3390/biology11081229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 05/24/2023]
Abstract
Glycoproteins and glycolipids on the cell surfaces of vertebrates and higher invertebrates contain α-keto acid sugars called sialic acids, terminally attached to their glycan structures. The actual level of sialylation, regulated through enzymatic removal of the latter ones by NEU enzymes, highly affects protein-protein, cell-matrix and cell-cell interactions. Thus, their regulatory features affect a large number of different cell types, including those of the immune system. Research regarding NEUs within heart and vessels provides new insights of their involvement in the development of cardiovascular pathologies and identifies mechanisms on how inhibiting NEU enzymes can have a beneficial effect on cardiac remodelling and on a number of different cardiac diseases including CMs and atherosclerosis. In this regard, a multitude of clinical studies demonstrated the potential of N-acetylneuraminic acid (Neu5Ac) to serve as a biomarker following cardiac diseases. Anti-influenza drugs i.e., zanamivir and oseltamivir are viral NEU inhibitors, thus, they block the enzymatic activity of NEUs. When considering the improvement in cardiac function in several different cardiac disease animal models, which results from NEU reduction, the inhibition of NEU enzymes provides a new potential therapeutic treatment strategy to treat cardiac inflammatory pathologies, and thus, administrate cardioprotection.
Collapse
|
35
|
Lemke G, Huang Y. The dense-core plaques of Alzheimer's disease are granulomas. J Exp Med 2022; 219:213305. [PMID: 35731195 PMCID: PMC9225945 DOI: 10.1084/jem.20212477] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/13/2022] [Accepted: 06/07/2022] [Indexed: 12/19/2022] Open
Abstract
Dense-core plaques, whose centers contain highly polymerized and compacted aggregates of amyloid β peptides, are one of the two defining histopathological features of Alzheimer's disease. Recent findings indicate that these plaques do not form spontaneously but are instead constructed by microglia, the tissue macrophages of the central nervous system. We discuss cellular, structural, functional, and gene expression criteria by which the microglial assembly of dense-core plaques in the Alzheimer's brain parallels the construction of granulomas by macrophages in other settings. We compare the genesis of these plaques to the macrophage assembly of mycobacterial granulomas, the defining histopathological features of tuberculosis. We suggest that if dense-core plaques are indeed granulomas, their simple disassembly may be contraindicated as an Alzheimer's therapy.
Collapse
Affiliation(s)
- Greg Lemke
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA.,Immunobiology and Microbial Pathogenesis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA
| | - Youtong Huang
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA
| |
Collapse
|
36
|
Doostkam A, Malekmakan L, Hosseinpour A, Janfeshan S, Roozbeh J, Masjedi F. Sialic acid: an attractive biomarker with promising biomedical applications. ASIAN BIOMED 2022; 16:153-167. [PMID: 37551166 PMCID: PMC10321195 DOI: 10.2478/abm-2022-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This broad, narrative review highlights the roles of sialic acids as acidic sugars found on cellular membranes. The role of sialic acids in cellular communication and development has been well established. Recently, attention has turned to the fundamental role of sialic acids in many diseases, including viral infections, cardiovascular diseases, neurological disorders, diabetic nephropathy, and malignancies. Sialic acid may be a target for developing new drugs to treat various cancers and inflammatory processes. We recommend the routine measurement of serum sialic acid as a sensitive inflammatory marker in various diseases.
Collapse
Affiliation(s)
- Aida Doostkam
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Leila Malekmakan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Alireza Hosseinpour
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz7134853185, Iran
| | - Sahar Janfeshan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Jamshid Roozbeh
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| | - Fatemeh Masjedi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz7193635899, Iran
| |
Collapse
|
37
|
Alteration of the neuronal and glial cell profiles in Neu1-deficient zebrafish. Glycoconj J 2022; 39:499-512. [PMID: 35877057 DOI: 10.1007/s10719-022-10074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/04/2022]
Abstract
Neu1 is a glycosidase that releases sialic acids from the non-reducing ends of glycoconjugates, and its enzymatic properties are conserved among vertebrates. Recently, Neu1-KO zebrafish were generated using genome editing technology, and the KO fish showed abnormal emotional behavior, such as low schooling, low aggressiveness, and excess exploratory behavior, accompanied by the downregulation of anxiety-related genes. To examine the alteration of neuronal and glial cells in Neu1-KO zebrafish, we analyzed the molecular profiles in the zebrafish brain, focusing on the midbrain and telencephalon. Using immunohistochemistry, we found that signals of Maackia amurensis (MAM) lectin that recognizes Sia α2-3 linked glycoconjugates were highly increased in Neu1-KO zebrafish brains, accompanied by an increase in Lamp1a. Neu1-KO zebrafish suppressed the gene expression of AMPA-type glutamate receptors such as gria1a, gria2a, and gria3b, and vesicular glutamate transporter 1. Additionally, Neu1-KO zebrafish induced the hyperactivation of astrocytes accompanied by an increase in Gfap and phosphorylated ERK levels, while the mRNA levels of astrocyte glutamate transporters (eaat1a, eaat1c, and eaat2) were downregulated. The mRNA levels of sypb and ho1b, which are markers of synaptic plasticity, were also suppressed by Neu1 deficiency. Abnormal activity of microglia was also revealed by IHC, and the expressions of iNOS and IL-1β, an inflammatory cytokine, were increased in Neu1-KO zebrafish. Furthermore, drastic neuronal degeneration was detected in Neu1-KO zebrafish using Fluoro-Jade B staining. Collectively, the neuronal and glial abnormalities in Neu1-KO zebrafish may be caused by changes in the excitatory neurotransmitter glutamate and involved in the emotional abnormalities.
Collapse
|
38
|
Khan A, Sergi CM. NEU1—A Unique Therapeutic Target for Alzheimer’s Disease. Front Pharmacol 2022; 13:902259. [PMID: 35847014 PMCID: PMC9277458 DOI: 10.3389/fphar.2022.902259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Neuraminidase 1 (NEU1) is considered to be the most abundant and ubiquitous mammalian enzyme, with a broad tissue distribution. It plays a crucial role in a variety of cellular mechanisms. The deficiency of NEU1 has been implicated in various pathological manifestations of sialidosis and neurodegeneration. Thus, it is a novel therapeutic target for neurodegenerative changes in the Alzheimer’s brain. However, to manipulate NEU1 as a therapeutic target, it is imperative to understand that, although NEU1 is commonly known for its lysosomal catabolic function, it is also involved in other pathways. NEU1 is involved in immune response modulation, elastic fiber assembly modulation, insulin signaling, and cell proliferation. In recent years, our knowledge of NEU1 has continued to grow, yet, at the present moment, current data is still limited. In addition, the unique biochemical properties of NEU1 make it challenging to target it as an effective therapeutic option for sialidosis, which is a rare disease but has an enormous patient burden. However, the fact that NEU1 has been linked to the pathology of Alzheimer’s disease, which is rapidly growing worldwide, makes it more relevant to be studied and explored. In the present study, the authors have discussed various cellular mechanisms involving NEU1 and how they are relevant to sialidosis and Alzheimer’s disease.
Collapse
Affiliation(s)
- Aiza Khan
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Consolato M. Sergi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
- Division of Anatomic Pathology, Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
- *Correspondence: Consolato M. Sergi,
| |
Collapse
|
39
|
Ganapathy S. R, Levová K, Kotačková L, Trnka J, Zogala D, Rusz J, Zima T, Devos D, Šonka K, Růžička E, Kalousová M, Dušek P. Increased Transferrin Sialylation Predicts Phenoconversion in Isolated REM Sleep Behavior Disorder. Mov Disord 2022; 37:983-992. [PMID: 35128728 PMCID: PMC9305135 DOI: 10.1002/mds.28942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Sialic acid-protein interactions are involved in regulating central nervous system immunity; therefore, derangements in sialylation could be involved in neurodegeneration. OBJECTIVES We evaluate the differences in serum transferrin sialylation in prodromal and early-stage Parkinson's disease (PD), its relation to substantia nigra degeneration, and the risk of phenoconversion to manifest disease. METHODS Sixty treatment-naive PD patients; 72 polysomnography-confirmed isolated rapid eye movement sleep behavior disorder (iRBD) patients, that is, patients with prodromal synucleinopathy; and 46 healthy volunteers aged ≥45 years and drinking ≤60 standard drinks per month were included. The proportion of serum low-sialylated, carbohydrate-deficient transferrin (CDT) isoforms was assessed using high-performance liquid chromatography, and the values were adjusted for alcohol intake (CDTadj ). Dopamine transporter single-photon emission computed tomography (DaT-SPECT) imaging was performed. In iRBD, phenoconversion risk of DaT-SPECT and CDTadj was evaluated using Cox regression adjusted for age and sex. RESULTS Median CDTadj was lower in PD (1.1 [interquartile range: 1.0-1.3]%) compared to controls (1.2 [1.1-1.6]%) (P = 0.001). In iRBD, median CDTadj was lower in subjects with abnormal (1.1 [0.9-1.3]%) than normal (1.3 [1.2-1.6]%) DaT-SPECT (P = 0.005). After a median 44-month follow-up, 20% of iRBD patients progressed to a manifest disease. Although iRBD converters and nonconverters did not significantly differ in CDTadj levels (P = 0.189), low CDTadj increased the risk of phenoconversion with hazard ratio 3.2 (P = 0.045) but did not refine the phenoconversion risk associated with abnormal DaT-SPECT yielding hazard ratio 15.8 (P < 0.001). CONCLUSIONS Decreased serum CDTadj is associated with substantia nigra degeneration in synucleinopathies. iRBD patients with low CDTadj are more likely to phenoconvert to manifest disease. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Ranjani Ganapathy S.
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Kateřina Levová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Lenka Kotačková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Jiří Trnka
- Institute of Nuclear Medicine, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - David Zogala
- Institute of Nuclear Medicine, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Jan Rusz
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
- Department of Circuit Theory, Faculty of Electrical EngineeringCzech Technical University in PraguePragueCzech Republic
| | - Tomáš Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - David Devos
- Department of Medical Pharmacology, Expert Center for Parkinson, CHU‐Lille, Lille Neuroscience and Cognition, Inserm, UMR‐S1172, LICEND, NS‐Park NetworkUniversity of LilleLilleFrance
| | - Karel Šonka
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Evžen Růžička
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Marta Kalousová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| | - Petr Dušek
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PraguePragueCzech Republic
| |
Collapse
|
40
|
Xiang J, Meng X, Zhao Y, Wu FX, Li M. HyMM: hybrid method for disease-gene prediction by integrating multiscale module structure. Brief Bioinform 2022; 23:6547263. [PMID: 35275996 DOI: 10.1093/bib/bbac072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/18/2022] [Accepted: 02/13/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Identifying disease-related genes is an important issue in computational biology. Module structure widely exists in biomolecule networks, and complex diseases are usually thought to be caused by perturbations of local neighborhoods in the networks, which can provide useful insights for the study of disease-related genes. However, the mining and effective utilization of the module structure is still challenging in such issues as a disease gene prediction. RESULTS We propose a hybrid disease-gene prediction method integrating multiscale module structure (HyMM), which can utilize multiscale information from local to global structure to more effectively predict disease-related genes. HyMM extracts module partitions from local to global scales by multiscale modularity optimization with exponential sampling, and estimates the disease relatedness of genes in partitions by the abundance of disease-related genes within modules. Then, a probabilistic model for integration of gene rankings is designed in order to integrate multiple predictions derived from multiscale module partitions and network propagation, and a parameter estimation strategy based on functional information is proposed to further enhance HyMM's predictive power. By a series of experiments, we reveal the importance of module partitions at different scales, and verify the stable and good performance of HyMM compared with eight other state-of-the-arts and its further performance improvement derived from the parameter estimation. CONCLUSIONS The results confirm that HyMM is an effective framework for integrating multiscale module structure to enhance the ability to predict disease-related genes, which may provide useful insights for the study of the multiscale module structure and its application in such issues as a disease-gene prediction.
Collapse
Affiliation(s)
- Ju Xiang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China; Department of Basic Medical Sciences & Academician Workstation, Changsha Medical University, Changsha, Hunan 410219, China
| | - Xiangmao Meng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yichao Zhao
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Fang-Xiang Wu
- Division of Biomedical Engineering and Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
41
|
The functional mechanism of bone marrow-derived mesenchymal stem cells in the treatment of animal models with Alzheimer's disease: crosstalk between autophagy and apoptosis. Stem Cell Res Ther 2022; 13:90. [PMID: 35241159 PMCID: PMC8895531 DOI: 10.1186/s13287-022-02765-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
The transplantation of bone marrow-derived mesenchymal stem cells (BMMSCs) alleviates neuropathology and improves cognitive deficits in animal models with Alzheimer's disease. However, the underlying mechanism remains undefined. Based on meta-analysis and comprehensive review, high-profile studies support the theory that transplanted BMMSCs activate autophagy, as evidenced by the expression levels of signal molecules such as Beclin-1, Atg5, LC3-II, and mTOR. Functional autophagy mitigates neuronal apoptosis, which is reflected by the alterations of IAPs, Bcl-2, caspase-3, and so forth. Moreover, the transplantation of BMMSCs can decrease aberrant amyloid-beta peptides as well as tau aggregates, inhibit neuroinflammation, and stimulate synaptogenesis. There is a signal crosstalk between autophagy and apoptosis, which may be regulated to produce synergistic effect on the preconditioning of stem cells. Forasmuch, the therapeutic effect of transplanted BMMSCs can be enhanced by autophagy and/or apoptosis modulators.
Collapse
|
42
|
Bourguet E, Figurska S, Fra Czek MM. Human Neuraminidases: Structures and Stereoselective Inhibitors. J Med Chem 2022; 65:3002-3025. [PMID: 35170942 DOI: 10.1021/acs.jmedchem.1c01612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This Perspective describes the classification, structures, substrates, mechanisms of action, and implications of human neuraminidases (hNEUs) in various pathologies. Some inhibitors have been developed for each isoform, leading to more precise interactions with hNEUs. Although crystal structure data are available for NEU2, most of the findings are based on NEU1 inhibition, and limited information is available for other hNEUs. Therefore, the synthesis of new compounds would facilitate the enrichment of the arsenal of inhibitors to better understand the roles of hNEUs and their mechanisms of action. Nevertheless, due to the already known inhibitors of human neuraminidase enzymes, a structure-activity relationship is presented along with different approaches to inhibit these enzymes for the development of potent and selective inhibitors. Among the different emerging strategies, one is the inhibition of the dimerization of NEU1 or NEU3, and the second is the inhibition of certain receptors located close to hNEU.
Collapse
Affiliation(s)
- Erika Bourguet
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), CNRS UMR 7312, 51097 Reims, France
| | - Sylwia Figurska
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), CNRS UMR 7312, 51097 Reims, France.,Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Manuela Maria Fra Czek
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), CNRS UMR 7312, 51097 Reims, France.,Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
43
|
Whyte LS, Fourrier C, Hassiotis S, Lau AA, Trim PJ, Hein LK, Hattersley KJ, Bensalem J, Hopwood JJ, Hemsley KM, Sargeant TJ. Lysosomal gene Hexb displays haploinsufficiency in a knock-in mouse model of Alzheimer’s disease. IBRO Neurosci Rep 2022; 12:131-141. [PMID: 35146484 PMCID: PMC8819126 DOI: 10.1016/j.ibneur.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/29/2022] Open
Abstract
Lysosomal network abnormalities are an increasingly recognised feature of Alzheimer’s disease (AD), which appear early and are progressive in nature. Sandhoff disease and Tay-Sachs disease (neurological lysosomal storage diseases caused by mutations in genes that code for critical subunits of β-hexosaminidase) result in accumulation of amyloid-β (Aβ) and related proteolytic fragments in the brain. However, experiments that determine whether mutations in genes that code for β-hexosaminidase are risk factors for AD are currently lacking. To determine the relationship between β-hexosaminidase and AD, we investigated whether a heterozygous deletion of Hexb, the gene that encodes the beta subunit of β-hexosaminidase, modifies the behavioural phenotype and appearance of disease lesions in AppNL-G-F/NL-G-F(AppKI/KI) mice. AppKI/KI and Hexb+/- mice were crossed and evaluated in a behavioural test battery. Neuropathological hallmarks of AD and ganglioside levels in the brain were also examined. Heterozygosity of Hexb in AppKI/KI mice reduced learning flexibility during the Reversal Phase of the Morris water maze. Contrary to expectation, heterozygosity of Hexb caused a small but significant decrease in amyloid beta deposition and an increase in the microglial marker IBA1 that was region- and age-specific. Hexb heterozygosity caused detectable changes in the brain and in the behaviour of an AD model mouse, consistent with previous reports that described a biochemical relationship between HEXB and AD. This study reveals that the lysosomal enzyme gene Hexb is not haplosufficient in the mouse AD brain. The App NL-G-F Alzheimer mouse has lysosomal defects and stores ganglioside lipids. Heterozygous lysosomal Hexb did not drive amyloidosis in the App NL-G-F mouse. Heterozygous Hexb on an Alzheimer’s background reduced learning flexibility. Heterozygous Hexb on a wild-type mouse background produced hypoactivity.
Collapse
|
44
|
Razani E, Pourbagheri-Sigaroodi A, Safaroghli-Azar A, Zoghi A, Shanaki-Bavarsad M, Bashash D. The PI3K/Akt signaling axis in Alzheimer's disease: a valuable target to stimulate or suppress? Cell Stress Chaperones 2021; 26:871-887. [PMID: 34386944 PMCID: PMC8578535 DOI: 10.1007/s12192-021-01231-3] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/23/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Among the long list of age-related complications, Alzheimer's disease (AD) has the most dreadful impact on the quality of life due to its devastating effects on memory and cognitive abilities. Although a plausible correlation between the phosphatidylinositol 3-kinase (PI3K) signaling and different processes involved in neurodegeneration has been evidenced, few articles reviewed the task. The current review aims to unravel the mechanisms by which the PI3K pathway plays pro-survival roles in normal conditions, and also to discuss the original data obtained from international research laboratories on this topic. Responses to questions on how alterations of the PI3K/Akt signaling pathway affect Tau phosphorylation and the amyloid cascade are given. In addition, we provide a general overview of the association between oxidative stress, neuroinflammation, alterations of insulin signaling, and altered autophagy with aberrant activation of this axis in the AD brain. The last section provides a special focus on the therapeutic possibility of the PI3K/Akt/mTOR modulators, either categorized as chemicals or herbals, in AD. In conclusion, determining the correct timing for the administration of the drugs seems to be one of the most important factors in the success of these agents. Also, the role of the PI3K/Akt signaling axis in the progression or repression of AD widely depends on the context of the cells; generally speaking, while PI3K/Akt activation in neurons and neural stem cells is favorable, its activation in microglia cells may be harmful.
Collapse
Affiliation(s)
- Elham Razani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ava Safaroghli-Azar
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anahita Zoghi
- Department of Neurology, School of Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Shanaki-Bavarsad
- Institute of Neuroscience, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Salminen A, Kaarniranta K, Kauppinen A. Hypoxia/ischemia impairs CD33 (Siglec-3)/TREM2 signaling: Potential role in Alzheimer's pathogenesis. Neurochem Int 2021; 150:105186. [PMID: 34530055 DOI: 10.1016/j.neuint.2021.105186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/22/2022]
Abstract
Recent genetic and molecular studies have indicated that the innate immune system, especially microglia, have a crucial role in the accumulation of β-amyloid plaques in Alzheimer's disease (AD). In particular, the CD33 receptor, also called Siglec-3, inhibits the TREM2 receptor-induced phagocytic activity of microglia. CD33 receptors recognize the α2,3 and α2,6-linked sialic groups in tissue glycocalyx, especially sialylated gangliosides in human brain. The CD33 receptor triggers cell-type specific responses, e.g., in microglia, CD33 inhibits phagocytosis, whereas in natural killer cells, it inhibits the cytotoxic activity of the NKG2D receptor. Nonetheless, the regulation of the activity of CD33 receptor needs to be clarified. For example, it seems that hypoxia/ischemia, a potential cause of AD pathology, increases the expression of CD33 and its downstream target SHP-1, a tyrosine phosphatase which suppresses the phagocytosis driven by TREM2. Moreover, hypoxia/ischemia increases the deposition of sialylated gangliosides, e.g., GM1, GM2, GM3, and GD1, which are ligands for inhibitory CD33/Siglec-3 receptors. In addition, β-amyloid peptides bind to the sialylated gangliosides in raft-like clusters and subsequently these gangliosides act as seeds for the formation of β-amyloid plaques in AD pathology. It is known that senile plaques contain sialylated GM1, GM2, and GM3 gangliosides, i.e., the same species induced by hypoxia/ischemia treatment. Sialylated gangliosides in plaques might stimulate the CD33/Siglec-3 receptors of microglia and thus impede TREM2-driven phagocytosis. We propose that hypoxia/ischemia, e.g., via the accumulation of sialylated gangliosides, prevents the phagocytosis of β-amyloid deposits by inhibiting CD33/TREM2 signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, KYS, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
46
|
Dilna A, Deepak KV, Damodaran N, Kielkopf CS, Kagedal K, Ollinger K, Nath S. Amyloid-β induced membrane damage instigates tunneling nanotube-like conduits by p21-activated kinase dependent actin remodulation. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166246. [PMID: 34403739 DOI: 10.1016/j.bbadis.2021.166246] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/19/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) pathology progresses gradually via anatomically connected brain regions. Direct transfer of amyloid-β1-42 oligomers (oAβ) between connected neurons has been shown, however, the mechanism is not fully revealed. We observed formation of oAβ induced tunneling nanotubes (TNTs)-like nanoscaled f-actin containing membrane conduits, in differentially differentiated SH-SY5Y neuronal models. Time-lapse images showed that oAβ propagate from one cell to another via TNT-like structures. Preceding the formation of TNT-like conduits, we detected oAβ-induced plasma membrane (PM) damage and calcium-dependent repair through lysosomal-exocytosis, followed by massive endocytosis to re-establish the PM. Massive endocytosis was monitored by an influx of the membrane-staining dye TMA-DPH and PM damage was quantified by propidium iodide influx in the absence of Ca2+. The massive endocytosis eventually caused accumulation of internalized oAβ in Lamp1 positive multivesicular bodies/lysosomes via the actin cytoskeleton remodulating p21-activated kinase1 (PAK1) dependent endocytic pathway. Three-dimensional quantitative confocal imaging, structured illumination superresolution microscopy, and flowcytometry quantifications revealed that oAβ induces activation of phospho-PAK1, which modulates the formation of long stretched f-actin extensions between cells. Moreover, the formation of TNT-like conduits was inhibited by preventing PAK1-dependent internalization of oAβ using the small-molecule inhibitor IPA-3, a highly selective cell-permeable auto-regulatory inhibitor of PAK1. The present study reveals that the TNT-like conduits are probably instigated as a consequence of oAβ induced PM damage and repair process, followed by PAK1 dependent endocytosis and actin remodeling, probably to maintain cell surface expansion and/or membrane tension in equilibrium.
Collapse
Affiliation(s)
- Aysha Dilna
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India
| | - K V Deepak
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India
| | - Nandini Damodaran
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India
| | - Claudia S Kielkopf
- Experimental Pathology, Department of Biomedical and Clinical Sciences Linköping University, 581 85 Linköping, Sweden
| | - Katarina Kagedal
- Experimental Pathology, Department of Biomedical and Clinical Sciences Linköping University, 581 85 Linköping, Sweden
| | - Karin Ollinger
- Experimental Pathology, Department of Biomedical and Clinical Sciences Linköping University, 581 85 Linköping, Sweden
| | - Sangeeta Nath
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India.
| |
Collapse
|
47
|
Bowles WHD, Gloster TM. Sialidase and Sialyltransferase Inhibitors: Targeting Pathogenicity and Disease. Front Mol Biosci 2021; 8:705133. [PMID: 34395532 PMCID: PMC8358268 DOI: 10.3389/fmolb.2021.705133] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
Sialidases (SAs) and sialyltransferases (STs), the enzymes responsible for removing and adding sialic acid to other glycans, play essential roles in viruses, bacteria, parasites, and humans. Sialic acid is often the terminal sugar on glycans protruding from the cell surface in humans and is an important component for recognition and cell function. Pathogens have evolved to exploit this and use sialic acid to either “cloak” themselves, ensuring they remain undetected, or as a mechanism to enable release of virus progeny. The development of inhibitors against SAs and STs therefore provides the opportunity to target a range of diseases. Inhibitors targeting viral, bacterial, or parasitic enzymes can directly target their pathogenicity in humans. Excellent examples of this can be found with the anti-influenza drugs Zanamivir (Relenza™, GlaxoSmithKline) and Oseltamivir (Tamiflu™, Roche and Gilead), which have been used in the clinic for over two decades. However, the development of resistance against these drugs means there is an ongoing need for novel potent and specific inhibitors. Humans possess 20 STs and four SAs that play essential roles in cellular function, but have also been implicated in cancer progression, as glycans on many cancer cells are found to be hyper-sialylated. Whilst much remains unknown about how STs function in relation to disease, it is clear that specific inhibitors of them can serve both as tools to gain a better understanding of their activity and form the basis for development of anti-cancer drugs. Here we review the recent developments in the design of SA and ST inhibitors against pathogens and humans.
Collapse
Affiliation(s)
- William H D Bowles
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, St Andrews, United Kingdom
| | - Tracey M Gloster
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
48
|
Neu1 deficiency induces abnormal emotional behavior in zebrafish. Sci Rep 2021; 11:13477. [PMID: 34188220 PMCID: PMC8241872 DOI: 10.1038/s41598-021-92778-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
NEU1 sialidase hydrolyzes sialic acids from glycoconjugates in lysosomes. Deficiency of NEU1 causes sialidosis with symptoms including facial dysmorphism, bone dysplasia, and neurodegeneration. However, the effects of NEU1 deficiency on emotional activity have not been explored. Here, we conducted the behavioral analysis using Neu1-knockout zebrafish (Neu1-KO). Neu1-KO zebrafish showed normal swimming similar to wild-type zebrafish (WT), whereas shoaling was decreased and accompanied by greater inter-fish distance than WT zebrafish. The aggression test showed a reduced aggressive behavior in Neu1-KO zebrafish than in WT zebrafish. In the mirror and 3-chambers test, Neu1-KO zebrafish showed more interest toward the opponent in the mirror and multiple unfamiliar zebrafish, respectively, than WT zebrafish. Furthermore, Neu1-KO zebrafish also showed increased interaction with different fish species, whereas WT zebrafish avoided them. In the black-white preference test, Neu1-KO zebrafish showed an abnormal preference for the white region, whereas WT zebrafish preferred the black region. Neu1-KO zebrafish were characterized by a downregulation of the anxiety-related genes of the hypothalamic-pituitary-adrenal axis and upregulation of lamp1a, an activator of lysosomal exocytosis, with their brains accumulating several sphingoglycolipids. This study revealed that Neu1 deficiency caused abnormal emotional behavior in zebrafish, possibly due to neuronal dysfunction induced by lysosomal exocytosis.
Collapse
|
49
|
Seol B, Kim YD, Cho YS. Modeling Sialidosis with Neural Precursor Cells Derived from Patient-Derived Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22094386. [PMID: 33922276 PMCID: PMC8122832 DOI: 10.3390/ijms22094386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 01/16/2023] Open
Abstract
Sialidosis, caused by a genetic deficiency of the lysosomal sialidase gene (NEU1), is a systemic disease involving various tissues and organs, including the nervous system. Understanding the neurological dysfunction and pathology associated with sialidosis remains a challenge, partially due to the lack of a human model system. In this study, we have generated two types of induced pluripotent stem cells (iPSCs) with sialidosis-specific NEU1G227R and NEU1V275A/R347Q mutations (sialidosis-iPSCs), and further differentiated them into neural precursor cells (iNPCs). Characterization of NEU1G227R- and NEU1V275A/R347Q- mutated iNPCs derived from sialidosis-iPSCs (sialidosis-iNPCs) validated that sialidosis-iNPCs faithfully recapitulate key disease-specific phenotypes, including reduced NEU1 activity and impaired lysosomal and autophagic function. In particular, these cells showed defective differentiation into oligodendrocytes and astrocytes, while their neuronal differentiation was not notably affected. Importantly, we found that the phenotypic defects of sialidosis-iNPCs, such as impaired differentiation capacity, could be effectively rescued by the induction of autophagy with rapamycin. Our results demonstrate the first use of a sialidosis-iNPC model with NEU1G227R- and NEU1V275A/R347Q- mutation(s) to study the neurological defects of sialidosis, particularly those related to a defective autophagy-lysosome pathway, and may help accelerate the development of new drugs and therapeutics to combat sialidosis and other LSDs.
Collapse
Affiliation(s)
- Binna Seol
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (B.S.); (Y.-D.K.)
| | - Young-Dae Kim
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (B.S.); (Y.-D.K.)
| | - Yee Sook Cho
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (B.S.); (Y.-D.K.)
- Department of Bioscience, KRIBB School, University of Science & Technology, 113 Gwahak-ro, Yuseong-gu, Daejeon 34113, Korea
- Correspondence: ; Tel.: +82-42-860-4479; Fax: +82-42-860-4608
| |
Collapse
|
50
|
Ammendolia DA, Bement WM, Brumell JH. Plasma membrane integrity: implications for health and disease. BMC Biol 2021; 19:71. [PMID: 33849525 PMCID: PMC8042475 DOI: 10.1186/s12915-021-00972-y] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma membrane integrity is essential for cellular homeostasis. In vivo, cells experience plasma membrane damage from a multitude of stressors in the extra- and intra-cellular environment. To avoid lethal consequences, cells are equipped with repair pathways to restore membrane integrity. Here, we assess plasma membrane damage and repair from a whole-body perspective. We highlight the role of tissue-specific stressors in health and disease and examine membrane repair pathways across diverse cell types. Furthermore, we outline the impact of genetic and environmental factors on plasma membrane integrity and how these contribute to disease pathogenesis in different tissues.
Collapse
Affiliation(s)
- Dustin A Ammendolia
- Cell Biology Program, Hospital for Sick Children, 686 Bay Street PGCRL, Toronto, ON, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - William M Bement
- Center for Quantitative Cell Imaging and Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, 686 Bay Street PGCRL, Toronto, ON, M5G 0A4, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,SickKids IBD Centre, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|