Next Article in Journal
Effects of Reversal of Hypotension on Cerebral Microcirculation and Metabolism in Experimental Sepsis
Previous Article in Journal
Oncology Drug Repurposing for Sepsis Treatment
Previous Article in Special Issue
Surgical Management of Valvular Heart Disease in Mucopolysaccharidoses: A Review of Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Molecular Mechanisms in Lysosomal Storage Diseases: From Pathogenesis to Therapeutic Strategies

by
Valeria De Pasquale
1,*,
Melania Scarcella
2 and
Luigi Michele Pavone
2,*
1
Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80127 Naples, Italy
2
Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
*
Authors to whom correspondence should be addressed.
Submission received: 6 April 2022 / Accepted: 14 April 2022 / Published: 17 April 2022
Lysosomal storage diseases (LSDs) are a group of metabolic diseases caused by inborn mutations of lysosomal enzymes, which lead to lysosome substrate accumulation in various cell types. As a result, a complex variety of pathogenic cascades are triggered, resulting in a clinical profile characterized by multisystemic involvement [1]. Indeed, LSDs are featured by a broad spectrum of clinical signs depending on both substrate and site of accumulation [2]. Among common clinical manifestations, nervous system (NS) implication is one of the most frequent signs and is shared among almost all types of LSDs [2].
Although the biochemical nature of LSDs was first revealed in 1934 [3], it is only over the last two decades that a series of studies have contributed to the understanding of the pathogenic mechanisms of such diseases [4,5,6,7,8,9,10]. These studies have disclosed the important role of the altered autophagy flux [4] and mitochondrial function [5], which, along with the stimulation of inflammatory response and immune abnormalities [6], appear to represent a common feature of many LSDs. In addition, the interplay between the accumulation/mislocalization of substrates and the alteration of many signaling pathways is also gaining attention in the context of LSDs [7,8,9,10]. However, despite advances that have been made in the treatment of these diseases, current therapies are available for only a subset of them, most of which only aim to ameliorate disease symptoms [11,12,13,14,15,16,17].
This Special Issue provides a comprehensive view of the molecular aspects of various LSDs. Our goal is to collect state-of-the-art research on LSDs and their pathomechanisms, with a focus on recent discoveries that have been made on the NS involvement, such as the association between LSDs and neurodegenerative disorders, or the use of neurological LSDs models for pathogenesis investigation and drug discovery.
Clarke et al. provided interesting insights on the interplay between LSDs and protein aggregation in neurodegenerative disorders. More specifically, they analyzed seven LSDs (Gaucher, Fabry, Sandhoff, Niemann–Pick A, Hurler, Pompe, and Niemann–Pick C) murine models and found evidence of neuroinflammation and α-synuclein, both reminiscent of α-synucleinopathies such as Parkinson’s disease [18].
Moreover, the role of lysosomal and endosomal dysfunction in the pathophysiology of neurodegenerative disorders is well known. Bécot et al. provided a review that focuses on the association between endosomal system impairment and the generation of amyloid plaques in Alzheimer’s disease (AD). They highlighted how AD-related dysfunction of the endosomal system, which diverts from a degradative to a secretory function, can promote the formation of exosome and exocytosis of pathogenic amyloid species [19].
Another association between neurodegenerative disorders and lysosomal dysfunction was reported by Bicchi et al. They reported lysosomal engulfment and autophagy impairment in primary fibroblast of transgenic mice expressing a mutant form of superoxide dismutase 1 (SOD1), a gene frequently mutated in amyotrophic lateral sclerosis type-1 (ALS1) [20].
Although NS involvement is the most severe consequence of the majority of LSDs, the impairment of other organs and tissues is very common. For instance, it is estimated that 60–90% of mucopolysaccharidosis (MPS) patients, a type of LSD characterized by glycosaminoglycans (GAGs) accumulation, suffer from cardiac valve dysfunctions [21,22]. Moreover, deposition of GAGs can also occur in the vascular smooth muscle and in cardiac muscle cells. As a result, MPS is frequently associated with heart diseases such as myocardium fibrosis and coronary heart disease [23,24,25].
Additionally, a large screening study performed on kidney transplantation recipients highlighted the importance of the accurate and early diagnosis of the Fabry disease, an LSD caused by mutations in the enzyme alpha-galactosidase A (GLA), with its most life-threatening clinical sign represented by end-stage kidney failure [26].
During the last few decades, large efforts have been made to set up reproducible models that finely replicated the clinical features of LSDs patients. To this end, a widely used model organism is represented by Drosophila melanogaster: as reviewed by Rigon et al., given the evolutionary conservation of genes and pathways involved in several LSDs pathogenesis between the fruit fly and Homo sapiens, it can be used both to study molecular mechanisms underlying LSDs pathology and as a tool to new drugs and treatments discovery [27].
More recently, induced pluripotent stem cells (iPSCs) have been proven to be effective emerging tools for the study of some types of LSDs—such as metachromatic leukodystrophy (MLD) [28]. Nevertheless, factors such as the difficulty to yield somatic cells from patients, high cost, and uncertain physiological fidelity can significantly limit their employment [29]. In an attempt to overcome this limitation, Esmail et al., leveraging a deep machine-learning platform, developed artificially induced whole-brain organoids (aiWBO) as a model of MLD. aiWBO accurately simulated the phenotype of both wild-type and MLD nervous system cells. Importantly, the same research group demonstrated that such a model could also predict the efficacy of combined drug treatment that ameliorate the MLD disease profiles, rendering it eligible for new therapies discovery [30].
Altogether, the articles of this Special Issue contribute to enhancing the knowledge in the field of LSDs, with a focus on the nervous system involvement. In particular, the cited articles provide new insights on pathological mechanisms, setups of new study models, and the discovery of new therapies related to this group of metabolic diseases.

Funding

This research was supported by funds from the Cure Sanfilippo Foundation USA grant “Targeting Heparan Sulfate Proteoglycans as a Novel Therapeutic Strategy for Sanfilippo diseases” 2021–2024 to L.M.P.

Acknowledgments

The guest editor would like to thank all of the authors for their valuable contribution to this Special Issue and all of the reviewers for their helpful comments during the peer review process.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Filocamo, M.; Morrone, A. Lysosomal Storage Disorders: Molecular Basis and Laboratory Testing. Hum. Genom. 2011, 5, 156–169. [Google Scholar] [CrossRef] [PubMed]
  2. Sun, A. Lysosomal Storage Disease Overview. Ann. Transl. Med. 2018, 6, 476. [Google Scholar] [CrossRef] [PubMed]
  3. Capper, A.; Epstein, H.; Schless, R.A. Gaucher’s Disease. Report of a Case with Presentation of a Table Differentiating the Lipoid Disturbances. Am. J. Med. Sci. 1934, 188, 84. [Google Scholar] [CrossRef]
  4. Seranova, E.; Connolly, K.J.; Zatyka, M.; Rosenstock, T.R.; Barrett, T.; Tuxworth, R.I.; Sarkar, S. Dysregulation of Autophagy as a Common Mechanism in Lysosomal Storage Diseases. Essays Biochem. 2017, 61, 733–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. de la Mata, M.; Cotán, D.; Villanueva-Paz, M.; de Lavera, I.; Álvarez-Córdoba, M.; Luzón-Hidalgo, R.; Suárez-Rivero, J.M.; Tiscornia, G.; Oropesa-Ávila, M. Mitochondrial Dysfunction in Lysosomal Storage Disorders. Diseases 2016, 4, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Rigante, D.; Cipolla, C.; Basile, U.; Gulli, F.; Savastano, M.C. Overview of Immune Abnormalities in Lysosomal Storage Disorders. Immunol. Lett. 2017, 188, 79–85. [Google Scholar] [CrossRef]
  7. Platt, F.M.; d’Azzo, A.; Davidson, B.L.; Neufeld, E.F.; Tifft, C.J. Lysosomal Storage Diseases. Nat. Rev. Dis. Primers 2018, 4, 27. [Google Scholar] [CrossRef]
  8. Pshezhetsky, A.V. Lysosomal Storage of Heparan Sulfate Causes Mitochondrial Defects, Altered Autophagy, and Neuronal Death in the Mouse Model of Mucopolysaccharidosis III Type C. Autophagy 2016, 12, 1059–1060. [Google Scholar] [CrossRef] [Green Version]
  9. Fecarotta, S.; Gasperini, S.; Parenti, G. New Treatments for the Mucopolysaccharidoses: From Pathophysiology to Therapy. Ital. J. Pediatr. 2018, 44 (Suppl. 2), 135–143. [Google Scholar] [CrossRef]
  10. De Pasquale, V.; Pavone, L.M. Heparan Sulfate Proteoglycans: The Sweet Side of Development Turns Sour in Mucopolysaccharidoses. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 165539. [Google Scholar] [CrossRef]
  11. Marques, A.R.A.; Saftig, P. Lysosomal Storage Disorders-Challenges, Concepts and Avenues for Therapy: Beyond Rare Diseases. J. Cell Sci. 2019, 132, jcs221739. [Google Scholar] [CrossRef] [PubMed]
  12. De Pasquale, V.; Scerra, G.; Scarcella, M.; D’Agostino, M.; Pavone, L.M. Competitive Binding of Extracellular Accumulated Heparan Sulfate Reduces Lysosomal Storage Defects and Triggers Neuronal Differentiation in a Model of Mucopolysaccharidosis IIIB. Biochim. Biophys. Acta. Mol. Cell Res. 2021, 1868, 119113. [Google Scholar] [CrossRef] [PubMed]
  13. Parenti, G.; Medina, D.L.; Ballabio, A. The Rapidly Evolving View of Lysosomal Storage Diseases. EMBO Mol. Med. 2021, 13, e12836. [Google Scholar] [CrossRef] [PubMed]
  14. De Pasquale, V.; Sarogni, P.; Pistorio, V.; Cerulo, G.; Paladino, S.; Pavone, L.M. Targeting Heparan Sulfate Proteoglycans as a Novel Therapeutic Strategy for Mucopolysaccharidoses. Mol. Ther. Methods Clin. Dev. 2018, 10, 8–16. [Google Scholar] [CrossRef]
  15. De Pasquale, V.; Costanzo, M.; Siciliano, R.A.; Mazzeo, M.F.; Pistorio, V.; Bianchi, L.; Marchese, E.; Ruoppolo, M.; Pavone, L.M.; Caterino, M. Proteomic Analysis of Mucopolysaccharidosis IIIB Mouse Brain. Biomolecules 2020, 10, 355. [Google Scholar] [CrossRef] [Green Version]
  16. Schultz, M.L.; Tecedor, L.; Chang, M.; Davidson, B. Clarifying Lysosomal Storage Diseases. Trends Neurosci. 2011, 34, 401–410. [Google Scholar] [CrossRef] [Green Version]
  17. De Pasquale, V.; Caterino, M.; Costanzo, M.; Fedele, R.; Ruoppolo, M.; Pavone, L.M. Targeted Metabolomic Analysis of a Mucopolysaccharidosis IIIB Mouse Model Reveals an Imbalance of Branched-Chain Amino Acid and Fatty Acid Metabolism. Int. J. Mol. Sci. 2020, 21, 4211. [Google Scholar] [CrossRef]
  18. Clarke, J.; Kayatekin, C.; Viel, C.; Shihabuddin, L.; Sardi, S.P. Murine Models of Lysosomal Storage Diseases Exhibit Differences in Brain Protein Aggregation and Neuroinflammation. Biomedicines 2021, 9, 446. [Google Scholar] [CrossRef]
  19. Bécot, A.; Volgers, C.; van Niel, G. Transmissible Endosomal Intoxication: A Balance between Exosomes and Lysosomes at the Basis of Intercellular Amyloid Propagation. Biomedicines 2020, 8, 272. [Google Scholar] [CrossRef]
  20. Bicchi, I.; Morena, F.; Argentati, C.; Nodari, L.R.; Emiliani, C.; Gelati, M.; Vescovi, A.L.; Martino, S. Storage of Mutant Human SOD1 in Non-Neural Cells from the Type-1 Amyotrophic Lateral Sclerosis RatG93A Model Correlated with the Lysosomes’ Dysfunction. Biomedicines 2021, 9, 1080. [Google Scholar] [CrossRef]
  21. Belfiore, M.P.; Iacobellis, F.; Acampora, E.; Caiazza, M.; Rubino, M.; Monda, E.; Magaldi, M.R.; Tarallo, A.; Sasso, M.; De Pasquale, V.; et al. Aortopathies in Mouse Models of Pompe, Fabry and Mucopolysaccharidosis IIIB Lysosomal Storage Diseases. PLoS ONE 2020, 15, e0233050. [Google Scholar] [CrossRef] [PubMed]
  22. Bolourchi, M.; Renella, P.; Wang, R.Y. Aortic Root Dilatation in Mucopolysaccharidosis I-VII. Int. J. Mol. Sci. 2016, 17, 2004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Schiattarella, G.G.; Cerulo, G.; De Pasquale, V.; Cocchiaro, P.; Paciello, O.; Avallone, L.; Belfiore, M.P.; Iacobellis, F.; Di Napoli, D.; Magliulo, F.; et al. The Murine Model of Mucopolysaccharidosis IIIB Develops Cardiopathies over Time Leading to Heart Failure. PLoS ONE 2015, 10, e0131662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. De Pasquale, V.; Pezone, A.; Sarogni, P.; Tramontano, A.; Schiattarella, G.G.; Avvedimento, V.E.; Paladino, S.; Pavone, L.M. EGFR activation triggers cellular hypertrophy and lysosomal disease in NAGLU-depleted cardiomyoblasts, mimicking the hallmarks of mucopolysaccharidosis IIIB. Cell Death Dis. 2018, 9, 40. [Google Scholar] [CrossRef] [PubMed]
  25. Rosser, B.A.; Chan, C.; Hoschtitzky, A. Surgical Management of Valvular Heart Disease in Mucopolysaccharidoses: A Review of Literature. Biomedicines 2022, 10, 375. [Google Scholar] [CrossRef]
  26. Veroux, M.; Monte, I.P.; Rodolico, M.S.; Corona, D.; Bella, R.; Basile, A.; Palmucci, S.; Pistorio, M.L.; Lanza, G.; De Pasquale, C.; et al. Screening for Fabry Disease in Kidney Transplant Recipients: Experience of a Multidisciplinary Team. Biomedicines 2020, 8, 396. [Google Scholar] [CrossRef]
  27. Rigon, L.; De Filippis, C.; Napoli, B.; Tomanin, R.; Orso, G. Exploiting the Potential of Drosophila Models in Lysosomal Storage Disorders: Pathological Mechanisms and Drug Discovery. Biomedicines 2021, 9, 268. [Google Scholar] [CrossRef]
  28. Meneghini, V.; Frati, G.; Sala, D.; De Cicco, S.; Luciani, M.; Cavazzin, C.; Paulis, M.; Mentzen, W.; Morena, F.; Giannelli, S.; et al. Generation of Human Induced Pluripotent Stem Cell-Derived Bona Fide Neural Stem Cells for Ex Vivo Gene Therapy of Metachromatic Leukodystrophy. Stem. Cells Transl. Med. 2017, 6, 352–368. [Google Scholar] [CrossRef]
  29. Frati, G.; Luciani, M.; Meneghini, V.; De Cicco, S.; Ståhlman, M.; Blomqvist, M.; Grossi, S.; Filocamo, M.; Morena, F.; Menegon, A.; et al. Human IPSC-Based Models Highlight Defective Glial and Neuronal Differentiation from Neural Progenitor Cells in Metachromatic Leukodystrophy. Cell Death Dis. 2018, 9, 698. [Google Scholar] [CrossRef] [Green Version]
  30. Esmail, S.; Danter, W.R. Artificially Induced Pluripotent Stem Cell-Derived Whole-Brain Organoid for Modelling the Pathophysiology of Metachromatic Leukodystrophy and Drug Repurposing. Biomedicines 2021, 9, 440. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

De Pasquale, V.; Scarcella, M.; Pavone, L.M. Molecular Mechanisms in Lysosomal Storage Diseases: From Pathogenesis to Therapeutic Strategies. Biomedicines 2022, 10, 922. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10040922

AMA Style

De Pasquale V, Scarcella M, Pavone LM. Molecular Mechanisms in Lysosomal Storage Diseases: From Pathogenesis to Therapeutic Strategies. Biomedicines. 2022; 10(4):922. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10040922

Chicago/Turabian Style

De Pasquale, Valeria, Melania Scarcella, and Luigi Michele Pavone. 2022. "Molecular Mechanisms in Lysosomal Storage Diseases: From Pathogenesis to Therapeutic Strategies" Biomedicines 10, no. 4: 922. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10040922

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop