Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (48)

Search Parameters:
Keywords = lysosome enzyme transport

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 3082 KiB  
Article
Genetic Insights and Diagnostic Challenges in Highly Attenuated Lysosomal Storage Disorders
by Elena Urizar, Eamon P. McCarron, Chaitanya Gadepalli, Andrew Bentley, Peter Woolfson, Siying Lin, Christos Iosifidis, Andrew C. Browning, John Bassett, Udara D. Senarathne, Neluwa-Liyanage R. Indika, Heather J. Church, James A. Cooper, Jorge Menendez Lorenzo, Maria Elena Farrugia, Simon A. Jones, Graeme C. Black and Karolina M. Stepien
Genes 2025, 16(8), 915; https://doi.org/10.3390/genes16080915 (registering DOI) - 30 Jul 2025
Viewed by 730
Abstract
Background: Lysosomal storage diseases (LSDs) are a genetically and clinically heterogeneous group of inborn errors of metabolism caused by variants in genes encoding lysosomal hydrolases, membrane proteins, activator proteins, or transporters. These disease-causing variants lead to enzymatic deficiencies and the progressive accumulation of [...] Read more.
Background: Lysosomal storage diseases (LSDs) are a genetically and clinically heterogeneous group of inborn errors of metabolism caused by variants in genes encoding lysosomal hydrolases, membrane proteins, activator proteins, or transporters. These disease-causing variants lead to enzymatic deficiencies and the progressive accumulation of undegraded substrates within lysosomes, disrupting cellular function across multiple organ systems. While classical phenotypes typically manifest in infancy or early childhood with severe multisystem involvement, a combination of advances in molecular diagnostics [particularly next-generation sequencing (NGS)] and improved understanding of disease heterogeneity have enabled the identification of attenuated forms characterised by residual enzyme activity and later-onset presentations. These milder phenotypes often evade early recognition due to nonspecific or isolated symptoms, resulting in significant diagnostic delays and missed therapeutic opportunities. Objectives/Methods: This study characterises the clinical, biochemical, and molecular profiles of 10 adult patients diagnosed with LSDs, all representing attenuated forms, and discusses them alongside a narrative review. Results: Enzyme activity, molecular data, and phenotypic assessments are described to explore genotype–phenotype correlations and identify diagnostic challenges. Conclusions: These findings highlight the variable expressivity and organ involvement of attenuated LSDs and reinforce the importance of maintaining clinical suspicion in adults presenting with unexplained cardiovascular, neurological, ophthalmological, or musculoskeletal findings. Enhanced recognition of atypical presentations is critical to facilitate earlier diagnosis, guide management, and enable cascade testing for at-risk family members. Full article
(This article belongs to the Special Issue Molecular Basis and Genetics of Intellectual Disability)
Show Figures

Figure 1

21 pages, 1041 KiB  
Review
The Hidden Burden: Gastrointestinal Involvement in Lysosomal Storage Disorders
by Vincenza Gragnaniello, Chiara Cazzorla, Daniela Gueraldi, Andrea Puma, Christian Loro and Alberto B. Burlina
Metabolites 2025, 15(6), 361; https://doi.org/10.3390/metabo15060361 - 29 May 2025
Viewed by 842
Abstract
Background: Lysosomal storage disorders (LSDs) are rare inherited metabolic diseases characterized by defects in lysosomal enzyme function or membrane transport. These defects lead to substrate accumulation and multisystemic manifestations. This review focuses on gastrointestinal (GI) involvement in LSDs, which is a significant but [...] Read more.
Background: Lysosomal storage disorders (LSDs) are rare inherited metabolic diseases characterized by defects in lysosomal enzyme function or membrane transport. These defects lead to substrate accumulation and multisystemic manifestations. This review focuses on gastrointestinal (GI) involvement in LSDs, which is a significant but often overlooked aspect of these disorders. Methods: A comprehensive literature review was conducted to examine the pathophysiology, clinical presentation, diagnosis and management of GI manifestations in several LSDs, including Fabry disease, Gaucher disease, Pompe disease, Niemann–Pick disease type C, mucopolysaccharidoses and Wolman disease. Results: The pathogenesis of GI involvement in LSDs varies and encompasses substrate accumulation in enterocytes, mesenteric lymphadenopathy, mass effects, smooth muscle dysfunction, vasculopathy, neuropathy, inflammation and alterations to the microbiota. Clinical presentations range from non-specific symptoms, such as abdominal pain, diarrhea and malabsorption, to more severe complications, such as protein-losing enteropathy and inflammatory bowel disease. Diagnosis often requires a high level of suspicion, as GI symptoms may precede the diagnosis of the underlying LSD or be misattributed to more common conditions. Management strategies include disease-specific treatments, such as enzyme replacement therapy or substrate reduction therapy, as well as supportive care and targeted interventions for specific GI complications. Conclusions: This review highlights the importance of recognizing and properly managing GI manifestations in LSDs to improve patient outcomes and quality of life. It also emphasizes the need for further research to develop more effective treatments for life-threatening GI complications associated with these rare genetic disorders. Full article
(This article belongs to the Special Issue Research of Inborn Errors of Metabolism)
Show Figures

Figure 1

27 pages, 6414 KiB  
Article
Allosteric Modulation of GCase Enhances Lysosomal Activity and Reduces ER Stress in GCase-Related Disorders
by Ilaria Fregno, Natalia Pérez-Carmona, Mikhail Rudinskiy, Tatiana Soldà, Timothy J. Bergmann, Ana Ruano, Aida Delgado, Elena Cubero, Manolo Bellotto, Ana María García-Collazo and Maurizio Molinari
Int. J. Mol. Sci. 2025, 26(9), 4392; https://doi.org/10.3390/ijms26094392 - 6 May 2025
Viewed by 1349
Abstract
Variants in the GBA1 gene, encoding the lysosomal enzyme glucosylceramidase beta 1 (GCase), are linked to Parkinson’s disease (PD) and Gaucher disease (GD). Heterozygous variants increase PD risk, while homozygous variants lead to GD, a lysosomal storage disorder. Some GBA1 variants impair GCase [...] Read more.
Variants in the GBA1 gene, encoding the lysosomal enzyme glucosylceramidase beta 1 (GCase), are linked to Parkinson’s disease (PD) and Gaucher disease (GD). Heterozygous variants increase PD risk, while homozygous variants lead to GD, a lysosomal storage disorder. Some GBA1 variants impair GCase maturation in the endoplasmic reticulum, blocking lysosomal transport and causing glucosylceramide accumulation, which disrupts lysosomal function. This study explores therapeutic strategies to address these dysfunctions. Using Site-directed Enzyme Enhancement Therapy (SEE-Tx®), two structurally targeted allosteric regulators (STARs), GT-02287 and GT-02329, were developed and tested in GD patient-derived fibroblasts with relevant GCase variants. Treatment with GT-02287 and GT-02329 improved the folding of mutant GCase, protected the GCaseLeu444Pro variant from degradation, and facilitated the delivery of active GCase to lysosomes. This enhanced lysosomal function and reduced cellular stress. These findings validate the STARs’ mechanism of action and highlight their therapeutic potential for GCase-related disorders, including GD, PD, and Dementia with Lewy Bodies. Full article
(This article belongs to the Special Issue Molecular Research of Dystonia and Parkinson’s Disease)
Show Figures

Figure 1

22 pages, 6401 KiB  
Article
Heat Acclimation Enhances Brain Resilience to Acute Thermal Stress in Clarias fuscus by Modulating Cell Adhesion, Anti-Apoptotic Pathways, and Intracellular Degradation Mechanisms
by Yingyi Guan, Cunyu Duan, Xinyu Xie, Zhuoying Luo, Dayan Zhou, Yulei Zhang, Guangli Li, Yu Liao and Changxu Tian
Animals 2025, 15(9), 1220; https://doi.org/10.3390/ani15091220 - 25 Apr 2025
Viewed by 537
Abstract
Global climate change presents a significant challenge to aquatic ecosystems, with ectothermic fish being particularly sensitive to temperature fluctuations. The brain plays a crucial role in perceiving, regulating, and adapting to thermal changes, and its response to heat stress is crucial for survival. [...] Read more.
Global climate change presents a significant challenge to aquatic ecosystems, with ectothermic fish being particularly sensitive to temperature fluctuations. The brain plays a crucial role in perceiving, regulating, and adapting to thermal changes, and its response to heat stress is crucial for survival. However, the molecular mechanisms underlying heat stress and acclimation in fish brains remain poorly understood. This study aimed to investigate the adaptive mechanisms of Hong Kong catfish (Clarias fuscus) brains under heat acclimation and acute heat stress using transcriptome analysis. Fish were divided into two groups: a normal temperature group (NT, 26 °C for 90 days) and a heat-acclimated group (HT, 34 °C for 90 days), followed by acute heat stress (34 °C for 72 h) and recovery (26 °C for 72 h). Heat acclimation improved C. fuscus tolerance to acute heat stress, with faster gene responses and stronger neuroprotection. Key pathways enriched included cell adhesion and ECM-receptor interactions during recovery. Apoptosis regulation was balanced, with the HT group upregulating anti-apoptotic genes to mitigate neuronal cell death. Additionally, the lysosome–phagosome pathway was activated during recovery, facilitating the transport of lysosomal enzymes and the clearance of damaged cellular components, aiding neuronal repair. Ribosome biogenesis was suppressed under heat stress to conserve energy, but this suppression was less pronounced in the HT group. In summary, heat acclimation enhances neural protection in C. fuscus brains by promoting neuronal repair, suppressing apoptosis, and activating lysosomal pathways, thereby improving tolerance to acute heat stress. These findings offer a molecular basis for breeding heat-tolerant fish species in aquaculture, and deepen our understanding of thermal adaptation in aquatic animals amid global climate change. Full article
(This article belongs to the Section Animal Welfare)
Show Figures

Figure 1

9 pages, 2524 KiB  
Communication
Expression of Tailored α-N-Acetylglucosaminidase in Escherichia coli for Synthesizing Mannose-6-Phosphate on N-Linked Oligosaccharides of Lysosomal Enzymes
by Yunsong Cao and Wei Wang
Bioengineering 2025, 12(4), 425; https://doi.org/10.3390/bioengineering12040425 - 17 Apr 2025
Viewed by 490
Abstract
Lysosomal enzymes are synthesized as N-glycosylated glycoproteins with mannose-6-phosphate (M6P) moieties, which are responsible for their binding to M6P receptors and transporting to the lysosome. In the M6P biosynthetic pathway, a Man8GlcNAc2 glycoform is converted to M6P groups through two [...] Read more.
Lysosomal enzymes are synthesized as N-glycosylated glycoproteins with mannose-6-phosphate (M6P) moieties, which are responsible for their binding to M6P receptors and transporting to the lysosome. In the M6P biosynthetic pathway, a Man8GlcNAc2 glycoform is converted to M6P groups through two consecutive enzymatic reactions, including N-acetylglucosamine (GlcNAc)-1-phosphotransferase (GNPT), transferring GlcNAc-1-phosphate from UDP-GlcNAc to the C6 hydroxyl groups of mannose residues, and then, removal of the covering GlcNAc moiety from the GlcNAc-P-mannose phosphodiester was carried out using an α-N-acetylglucosaminidase (referred to as ‘uncovering enzyme’, UCE) in the trans-Golgi network (TGN). Here, we expressed differently tailored versions of the UCE, including four truncated variants, in Escherichia coli. The four variants with the signal peptide, transmembrane domain, propiece and cytoplasmic tail truncated, respectively, were purified by affinity chromatography, and their enzymatic activities were assayed using a UDP-Glo kit. By fusing a maltose-binding protein (MBP) in the N-terminus of the UCE variants, the fusion proteins could be soluble when expressed in E. coli. The highest concentration of the purified enzyme was 80.5 mg/L of fermentation broth. Furthermore, the UCE with the core catalytic domain exhibited the highest uncovering activity. Full article
(This article belongs to the Section Biochemical Engineering)
Show Figures

Graphical abstract

28 pages, 10013 KiB  
Article
A TMT-Based Proteomic Analysis of Osmoregulation in the Gills of Oreochromis mossambicus Exposed to Three Osmotic Stresses
by Huanhuan Su, Dongmei Ma, Jiajia Fan, Zaixuan Zhong, Yuanyuan Tian and Huaping Zhu
Int. J. Mol. Sci. 2025, 26(6), 2791; https://doi.org/10.3390/ijms26062791 - 20 Mar 2025
Viewed by 2889
Abstract
Salinity and alkalinity are critical environmental factors that affect fish physiology and ability to survive. Oreochromis mossambicus is a euryhaline species that can endure a wide range of salinities and has the potential to serve as a valuable model animal for environmental science. [...] Read more.
Salinity and alkalinity are critical environmental factors that affect fish physiology and ability to survive. Oreochromis mossambicus is a euryhaline species that can endure a wide range of salinities and has the potential to serve as a valuable model animal for environmental science. In order to detect the histomorphological changes, antioxidant enzymes, and proteomic responses of O. mossambicus to different osmotic stresses, O. mossambicus was subjected to salinity stress (25 g/L, S_S), alkalinity stress (4 g/L, A_S), saline–alkalinity stress (salinity: 25 g/L, alkalinity: 4 g/L, SA_S), and freshwater (the control group; C_S). The histomorphological and antioxidant enzyme results indicated that salinity, alkalinity, and saline–alkalinity stresses have different degrees of damage and effects on the gills and liver of O. mossambicus. Compared with the control, 83, 187, and 177 differentially expressed proteins (DEPs) were identified in the salinity, alkalinity, and saline–alkalinity stresses, respectively. The obtained DEPs can be summarized into four categories: ion transport channels or proteins, energy synthesis and metabolism, immunity, and apoptosis. The KEGG enrichment results indicated that DNA replication and repair were significantly enriched in the salinity stress group. Lysosomes and oxidative phosphorylation were considerably enriched in the alkalinity stress group. Comparatively, the three most important enriched pathways in the saline–alkalinity stress group were Parkinson’s disease, Alzheimer’s disease, and Huntington’s disease. The findings of this investigation yield robust empirical evidence elucidating osmoregulatory mechanisms and adaptive biological responses in euryhaline teleost, thereby establishing a scientific foundation for the cultivation and genomic exploration of high-salinity-tolerant teleost species. This advancement facilitates the sustainable exploitation of saline–alkaline aquatic ecosystems while contributing to the optimization of piscicultural practices in hypersaline environments. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

19 pages, 3698 KiB  
Article
Synthesis and Characterization of Memantine-Loaded Niosomes for Enhanced Alzheimer’s Disease Targeting
by Hasan Turkez, Sena Oner, Ozge Caglar Yıldırım, Mehmet Enes Arslan, Marilisa Pia Dimmito, Çigdem Yuce Kahraman, Lisa Marinelli, Erdal Sonmez, Özlem Kiki, Abdulgani Tatar, Ivana Cacciatore, Antonio Di Stefano and Adil Mardinoglu
Pharmaceutics 2025, 17(2), 267; https://doi.org/10.3390/pharmaceutics17020267 - 17 Feb 2025
Viewed by 1251
Abstract
Background/Objectives: Over the past 25 years, numerous biological molecules, like recombinant lysosomal enzymes, neurotrophins, receptors, and therapeutic antibodies, have been tested in clinical trials for neurological diseases. However, achieving significant success in clinical applications has remained elusive. A primary challenge has been the [...] Read more.
Background/Objectives: Over the past 25 years, numerous biological molecules, like recombinant lysosomal enzymes, neurotrophins, receptors, and therapeutic antibodies, have been tested in clinical trials for neurological diseases. However, achieving significant success in clinical applications has remained elusive. A primary challenge has been the inability of these molecules to traverse the blood–brain barrier (BBB). Recognizing this hurdle, our study aimed to utilize niosomes as delivery vehicles, leveraging the “molecular Trojan horse” technology, to enhance the transport of molecules across the BBB. Methods: Previously synthesized memantine derivatives (MP1–4) were encapsulated into niosomes for improved BBB permeability, hypothesizing that this approach could minimize peripheral drug toxicity while ensuring targeted brain delivery. Using the human neuroblastoma (SH-SY5Y) cell line differentiated into neuron-like structures with retinoic acid and then exposed to amyloid beta 1–42 peptide, we established an in vitro Alzheimer’s disease (AD) model. In this model, the potential usability of MP1–4 was assessed through viability tests (MTT) and toxicological response analysis. The niosomes’ particle size and morphological structures were characterized using scanning electron microscopy (SEM), with their loading and release capacities determined via UV spectroscopy. Crucially, the ability of the niosomes to cross the BBB and their potential anti-Alzheimer efficacy were analyzed in an in vitro transwell system with endothelial cells. Results: The niosomal formulations demonstrated effective drug encapsulation (encapsulation efficiency: 85.3% ± 2.7%), controlled release (72 h release: 38.5% ± 1.2%), and stable morphology (PDI: 0.22 ± 0.03, zeta potential: −31.4 ± 1.5 mV). Among the derivatives, MP1, MP2, and MP4 exhibited significant neuroprotective effects, enhancing cell viability by approximately 40% (p < 0.05) in the presence of Aβ1-42 at a concentration of 47 µg/mL. The niosomal delivery system improved BBB permeability by 2.5-fold compared to free drug derivatives, as confirmed using an in vitro bEnd.3 cell model. Conclusions: Memantine-loaded niosomes provide a promising platform for overcoming BBB limitations and enhancing the therapeutic efficacy of Alzheimer’s disease treatments. This study highlights the potential of nanotechnology-based delivery systems in developing targeted therapies for neurodegenerative diseases. Further in vivo studies are warranted to validate these findings and explore clinical applications. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

16 pages, 270 KiB  
Article
Effects of Dietary Copper Sources and Levels on Liver Copper Metabolism and the Expression of Transporters in Growing Pigs
by Rui Sun, Meng Li, Tianrui Zhang, Wenyan Yang and Lianyu Yang
Animals 2025, 15(4), 526; https://doi.org/10.3390/ani15040526 - 12 Feb 2025
Viewed by 963
Abstract
Research on the effects of organic and inorganic Cu sources on metabolic processes and mechanisms in pigs is lacking. This study investigated the effects of different copper (Cu) sources and levels on hepatic Cu metabolism and transporter factors in growing pigs. Sixty healthy [...] Read more.
Research on the effects of organic and inorganic Cu sources on metabolic processes and mechanisms in pigs is lacking. This study investigated the effects of different copper (Cu) sources and levels on hepatic Cu metabolism and transporter factors in growing pigs. Sixty healthy piglets (initial body weight 14.00 ± 0.30 kg) were randomly divided into four groups with five replicates of three pigs each. Four diets (AM, AH, BM, and BH) had different Cu sources [Cu sulphate (CuSO4): A and Cu amino acids (Cu-AA): B] and levels [supplemented (120 mg/kg DM): M, supplemented (240 mg/kg DM): H]. The pre-feeding period was 7 days, followed by a 45-day feeding period. Slaughter and sample collection were carried out on the 46th day of the formal feeding period. Significant differences were considered at p < 0.05. The final weight and average daily gain (ADG) of growing pigs in the Cu-AA groups were significantly higher than those in the CuSO4 groups. Serum Cu increased with increasing Cu supplementation on days 20 and 40. Cu concentrations in muscle, liver, and liver subcellular organelles were higher in Cu-AA groups. In the CuSO4 groups, Cu concentrations were higher in kidneys and faeces. In Cu-AA groups, both the Cu concentrations in lysosomes and cytosol were higher, and the activities of cathepsin D (CTSD), β-glucosidase (BGL), and acid phosphatase (ACP) in lysosomes and cytoplasm were higher. Comparisons between groups showed that liver mRNA of copper transporter protein 1 (CTR1), ATPase copper-transporting beta (ATP7B), ceruloplasmin (CP), antioxidant protein 1 (ATOX1), and metallothionein (MT) was lower in the CuSO4 group than in the Cu-AA group, with the best performance at 120 mg/kg Cu. mRNAs for ATPase copper-transporting alpha (ATP7A), cytochrome c oxidase copper chaperone 17 (COX17), and copper chaperone for superoxide dismutase (CCS) showed a decreasing trend in the Cu-AA groups. Cu-AA is better for Cu deposition, enhances the utilisation of Cu, reduces Cu excretion, and promotes the expression of relevant enzymes and transporters in the liver. Full article
(This article belongs to the Special Issue Exploration of Sustainable Feed Resources and Pig Dietary Strategies)
16 pages, 2681 KiB  
Article
Toxicology Effects of Cadmium in Pomacea canaliculate: Accumulation, Oxidative Stress, Microbial Community, and Transcriptome Analysis
by Mingxin Qiu, Xiaoyang Bi, Yuanyang Liu, Huashou Li, Dongqin Li and Guikui Chen
Int. J. Mol. Sci. 2025, 26(2), 751; https://doi.org/10.3390/ijms26020751 - 17 Jan 2025
Cited by 4 | Viewed by 1330
Abstract
Cadmium (Cd) pollution poses an important problem, but limited information is available about the toxicology effects of Cd on freshwater invertebrates. We investigated the accumulation, oxidative stress, microbial community changes, and transcriptomic alterations in apple snails (Pomacea canaliculata) under Cd stress. The [...] Read more.
Cadmium (Cd) pollution poses an important problem, but limited information is available about the toxicology effects of Cd on freshwater invertebrates. We investigated the accumulation, oxidative stress, microbial community changes, and transcriptomic alterations in apple snails (Pomacea canaliculata) under Cd stress. The snails were exposed to the 10 μg/L Cd solution for 16 days, followed by a 16-day elimination period. Our results showed that the liver accumulated the highest Cd concentration (17.41 μg/g), followed by the kidneys (8.00 μg/g) and intestine-stomach (6.68 μg/g), highlighting these tissues as primary targets for Cd accumulation. During the elimination period, Cd concentrations decreased in all tissues, with the head-foot and shell exhibiting over 30% elimination rates. Cd stress also resulted in reduced activities of superoxide dismutase (SOD), catalase (CAT), and glutathione transferase (GST) compared to the control group. Notably, even after 16 days of depuration, the enzyme activities did not return to normal levels, indicating persistent toxicological effects. Cd exposure significantly reduced the diversity of gut microbiota in P. canaliculata. Moreover, transcriptome analysis identified differentially expressed genes (DEGs) primarily associated with lysosome function, motor proteins, protein processing in the endoplasmic reticulum, drug metabolism via cytochrome P450 (CYP450), arachidonic acid metabolism, and ECM–receptor interactions. These findings suggest that Cd stress predominantly disrupts cellular transport and metabolic processes. Overall, our study provides comprehensive insights into the toxicological impact of Cd on P. canaliculata and emphasizes the importance of understanding the mechanisms underlying Cd toxicity in aquatic organisms. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

17 pages, 4023 KiB  
Article
Investigating the Impact of the Parkinson’s-Associated GBA1 E326K Mutation on β-Glucocerebrosidase Dimerization and Interactome Dynamics Through an In Silico Approach
by Davide Pietrafesa, Alessia Casamassa, Barbara Benassi, Massimo Santoro, Massimo Marano, Claudia Consales, Jessica Rosati and Caterina Arcangeli
Int. J. Mol. Sci. 2024, 25(21), 11443; https://doi.org/10.3390/ijms252111443 - 24 Oct 2024
Cited by 2 | Viewed by 8438
Abstract
Heterozygous mutations or genetic variants in the GBA1 gene, which encodes for the β-glucocerebrosidase (GCase), a lysosomal hydrolase enzyme, may increase the risk of Parkinson’s disease (PD) onset. The heterozygous E326K form is one of the most common genetic risk factors for PD [...] Read more.
Heterozygous mutations or genetic variants in the GBA1 gene, which encodes for the β-glucocerebrosidase (GCase), a lysosomal hydrolase enzyme, may increase the risk of Parkinson’s disease (PD) onset. The heterozygous E326K form is one of the most common genetic risk factors for PD worldwide, but, to date, the underlying molecular mechanisms remain unclear. Here, we investigate the effect of the E326K on the structure, stability, dimerization process, and interaction mode with some proteins of the interactome of GCase using multiple molecular dynamics (MD) simulations at pH 5.5 and pH 7.0 to mimic the lysosomal and endoplasmic reticulum environments, respectively. The analysis of the MD trajectories highlights that the E326K mutation did not significantly alter the structural conformation of the catalytic dyad but significantly makes the structure of the dimeric complexes unstable, especially at lysosomal pH, potentially impacting the organization of the quaternary structure. Furthermore, the E326K mutation significantly impacts protein interactions by altering the binding mode with the activator Saposin C (SapC), reducing the binding affinity with the inhibitor α-Synuclein (α-Syn), and increasing the affinity for the Lysosomal integral membrane protein-2 (LIMP-2) transporter. Full article
Show Figures

Graphical abstract

18 pages, 921 KiB  
Review
Oncological Aspects of Lysosomal Storage Diseases
by Agnieszka Ługowska
Cells 2024, 13(19), 1664; https://doi.org/10.3390/cells13191664 - 8 Oct 2024
Cited by 2 | Viewed by 2096
Abstract
Lysosomal storage diseases (LSDs) are caused by the deficient activity of a lysosomal hydrolase or the lack of a functional membrane protein, transporter, activator, or other protein. Lysosomal enzymes break down macromolecular compounds, which contribute to metabolic homeostasis. Stored, undegraded materials have multiple [...] Read more.
Lysosomal storage diseases (LSDs) are caused by the deficient activity of a lysosomal hydrolase or the lack of a functional membrane protein, transporter, activator, or other protein. Lysosomal enzymes break down macromolecular compounds, which contribute to metabolic homeostasis. Stored, undegraded materials have multiple effects on cells that lead to the activation of autophagy and apoptosis, including the toxic effects of lyso-lipids, the disruption of intracellular Ca2+ ion homeostasis, the secondary storage of macromolecular compounds, the activation of signal transduction, apoptosis, inflammatory processes, deficiencies of intermediate compounds, and many other pathways. Clinical observations have shown that carriers of potentially pathogenic variants in LSD-associated genes and patients affected with some LSDs are at a higher risk of cancer, although the results of studies on the frequency of oncological diseases in LSD patients are controversial. Cancer is found in individuals affected with Gaucher disease, Fabry disease, Niemann-Pick type A and B diseases, alfa-mannosidosis, and sialidosis. Increased cancer prevalence has also been reported in carriers of a potentially pathogenic variant of an LSD gene, namely CLN3, SGSH, GUSB, NEU1, and, to a lesser extent, in other genes. In this review, LSDs in which oncological events can be observed are described. Full article
(This article belongs to the Collection The Lysosome in Cancer: From Pathogenesis to Therapy)
Show Figures

Figure 1

40 pages, 3082 KiB  
Systematic Review
Efficacy of Ivermectin, Chloroquine/Hydroxychloroquine, and Azithromycin in Managing COVID-19: A Systematic Review of Phase III Clinical Trials
by Nathália Mariana Santos Sansone, Matheus Negri Boschiero and Fernando Augusto Lima Marson
Biomedicines 2024, 12(10), 2206; https://doi.org/10.3390/biomedicines12102206 - 27 Sep 2024
Cited by 9 | Viewed by 11079
Abstract
Background: During the coronavirus disease (COVID)-19 pandemic several drugs were used to manage the patients mainly those with a severe phenotype. Potential drugs were used off-label and major concerns arose from their applicability to managing the health crisis highlighting the importance of clinical [...] Read more.
Background: During the coronavirus disease (COVID)-19 pandemic several drugs were used to manage the patients mainly those with a severe phenotype. Potential drugs were used off-label and major concerns arose from their applicability to managing the health crisis highlighting the importance of clinical trials. In this context, we described the mechanisms of the three repurposed drugs [Ivermectin-antiparasitic drug, Chloroquine/Hydroxychloroquine-antimalarial drugs, and Azithromycin-antimicrobial drug]; and, based on this description, the study evaluated the clinical efficacy of those drugs published in clinical trials. The use of these drugs reflects the period of uncertainty that marked the beginning of the COVID-19 pandemic, which made them a possible treatment for COVID-19. Methods: In our review, we evaluated phase III randomized controlled clinical trials (RCTs) that analyzed the efficacy of these drugs published from the COVID-19 pandemic onset to 2023. We included eight RCTs published for Ivermectin, 11 RCTs for Chloroquine/Hydroxychloroquine, and three RCTs for Azithromycin. The research question (PICOT) accounted for P—hospitalized patients with confirmed or suspected COVID-19; I—use of oral or intravenous Ivermectin OR Chloroquine/Hydroxychloroquine OR Azithromycin; C—placebo or no placebo (standard of care); O—mortality OR hospitalization OR viral clearance OR need for mechanical ventilation OR clinical improvement; and T—phase III RCTs. Results: While studying these drugs’ respective mechanisms of action, the reasons for which they were thought to be useful became apparent and are as follows: Ivermectin binds to insulin-like growth factor and prevents nuclear transportation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), therefore preventing cell entrance, induces apoptosis, and osmotic cell death and disrupts viral replication. Chloroquine/Hydroxychloroquine blocks the movement of SARS-CoV-2 from early endosomes to lysosomes inside the cell, also, this drug blocks the binding between SARS-CoV-2 and Angiotensin-Converting Enzyme (ACE)-2 inhibiting the interaction between the virus spike proteins and the cell membrane and this drug can also inhibit SARS-CoV-2 viral replication causing, ultimately, the reduction in viral infection as well as the potential to progression for a higher severity phenotype culminating with a higher chance of death. Azithromycin exerts a down-regulating effect on the inflammatory cascade, attenuating the excessive production of cytokines and inducing phagocytic activity, and acts interfering with the viral replication cycle. Ivermectin, when compared to standard care or placebo, did not reduce the disease severity, need for mechanical ventilation, need for intensive care unit, or in-hospital mortality. Only one study demonstrated that Ivermectin may improve viral clearance compared to placebo. Individuals who received Chloroquine/Hydroxychloroquine did not present a lower incidence of death, improved clinical status, or higher chance of respiratory deterioration compared to those who received usual care or placebo. Also, some studies demonstrated that Chloroquine/Hydroxychloroquine resulted in worse outcomes and side-effects included severe ones. Adding Azithromycin to a standard of care did not result in clinical improvement in hospitalized COVID-19 participants. In brief, COVID-19 was one of the deadliest pandemics in modern human history. Due to the potential health catastrophe caused by SARS-CoV-2, a global effort was made to evaluate treatments for COVID-19 to attenuate its impact on the human species. Unfortunately, several countries prematurely justified the emergency use of drugs that showed only in vitro effects against SARS-CoV-2, with a dearth of evidence supporting efficacy in humans. In this context, we reviewed the mechanisms of several drugs proposed to treat COVID-19, including Ivermectin, Chloroquine/Hydroxychloroquine, and Azithromycin, as well as the phase III clinical trials that evaluated the efficacy of these drugs for treating patients with this respiratory disease. Conclusions: As the main finding, although Ivermectin, Chloroquine/Hydroxychloroquine, and Azithromycin might have mechanistic effects against SARS-CoV-2 infection, most phase III clinical trials observed no treatment benefit in patients with COVID-19, underscoring the need for robust phase III clinical trials. Full article
Show Figures

Figure 1

18 pages, 2087 KiB  
Review
Ubiquitination and De-Ubiquitination in the Synthesis of Cow Milk Fat: Reality and Prospects
by Rui Gao, Yanni Wu, Yuhao Wang, Zhangping Yang, Yongjiang Mao, Yi Yang, Chunhua Yang and Zhi Chen
Molecules 2024, 29(17), 4093; https://doi.org/10.3390/molecules29174093 - 29 Aug 2024
Cited by 2 | Viewed by 1728
Abstract
Ubiquitination modifications permit the degradation of labelled target proteins with the assistance of proteasomes and lysosomes, which is the main protein degradation pathway in eukaryotic cells. Polyubiquitination modifications of proteins can also affect their functions. De-ubiquitinating enzymes reverse the process of ubiquitination via [...] Read more.
Ubiquitination modifications permit the degradation of labelled target proteins with the assistance of proteasomes and lysosomes, which is the main protein degradation pathway in eukaryotic cells. Polyubiquitination modifications of proteins can also affect their functions. De-ubiquitinating enzymes reverse the process of ubiquitination via cleavage of the ubiquitin molecule, which is known as a de-ubiquitination. It was demonstrated that ubiquitination and de-ubiquitination play key regulatory roles in fatty acid transport, de novo synthesis, and desaturation in dairy mammary epithelial cells. In addition, natural plant extracts, such as stigmasterol, promote milk fat synthesis in epithelial cells via the ubiquitination pathway. This paper reviews the current research on ubiquitination and de-ubiquitination in dairy milk fat production, with a view to providing a reference for subsequent research on milk fat and exploring new directions for the improvement of milk quality. Full article
Show Figures

Figure 1

11 pages, 1117 KiB  
Article
Intellectual Disabilities and Neurocognitive Impairment in Adult Patients with Inherited Metabolic Diseases: A UK Single Centre Experience
by John Warner-Levy, Adrian H. Heald, Daniel Hand, Reena Sharma, Rachel Thomasson and Karolina M. Stepien
Genes 2024, 15(7), 923; https://doi.org/10.3390/genes15070923 - 15 Jul 2024
Viewed by 2199
Abstract
Inherited metabolic diseases (IMDs) are a group of heterogeneous genetic disorders resulting in substrate accumulation, energy deficiency, or complex molecular defects due to the failure of specific molecules to act as enzymes, cofactors, transporters, or receptors in specific metabolic pathways. The pathophysiological changes [...] Read more.
Inherited metabolic diseases (IMDs) are a group of heterogeneous genetic disorders resulting in substrate accumulation, energy deficiency, or complex molecular defects due to the failure of specific molecules to act as enzymes, cofactors, transporters, or receptors in specific metabolic pathways. The pathophysiological changes seen in IMDs are sometimes associated with intellectual disability (ID) or neurocognitive decline, necessitating multidisciplinary input. We here describe our experience at one tertiary metabolic centre in the UK. We reviewed the case prevalence and existing service provision in one adult IMD service covering a multi-ethnic population of 10 million in North England. In our cohort of 2268 IMD patients, 1598 patients had general metabolic conditions (70.5%), and 670 had lysosomal storage disease/disorders (LSD)s (29.5%). The overall prevalence of ID and neurocognitive decline was found to be 15.7% (n = 357), with patients with LSDs accounting for 23.5% (n = 84) of affected patients. Given the prevalence of ID in adults with IMDs, access to multidisciplinary input from neuropsychology and neuropsychiatry services is important. Education of healthcare professionals to diagnose IMDs in patients with ID, in addition to neurocognitive and neuropsychiatric presentations, will avoid missed diagnoses of IMD and will have a positive effect on patient outcomes. Full article
(This article belongs to the Special Issue Molecular Genetics of Neurodevelopmental Disorders)
Show Figures

Figure 1

28 pages, 6607 KiB  
Article
Morquio A Syndrome: Identification of Differential Patterns of Molecular Pathway Interactions in Bone Lesions
by J. Victor. Álvarez, Susana B. Bravo, María Pilar Chantada-Vázquez, Carmen Pena, Cristóbal Colón, Shunji Tomatsu, Francisco J. Otero-Espinar and María L. Couce
Int. J. Mol. Sci. 2024, 25(6), 3232; https://doi.org/10.3390/ijms25063232 - 12 Mar 2024
Cited by 5 | Viewed by 2742
Abstract
Mucopolysaccharidosis type IVA (MPS IVA; Morquio A syndrome) is a rare autosomal recessive lysosomal storage disease (LSD) caused by deficiency of a hydrolase enzyme, N-acetylgalactosamine-6-sulfate sulfatase, and characterized clinically by mainly musculoskeletal manifestations. The mechanisms underlying bone involvement in humans are typically explored [...] Read more.
Mucopolysaccharidosis type IVA (MPS IVA; Morquio A syndrome) is a rare autosomal recessive lysosomal storage disease (LSD) caused by deficiency of a hydrolase enzyme, N-acetylgalactosamine-6-sulfate sulfatase, and characterized clinically by mainly musculoskeletal manifestations. The mechanisms underlying bone involvement in humans are typically explored using invasive techniques such as bone biopsy, which complicates analysis in humans. We compared bone proteomes using DDA and SWATH-MS in wild-type and MPS IVA knockout mice (UNT) to obtain mechanistic information about the disease. Our findings reveal over 1000 dysregulated proteins in knockout mice, including those implicated in oxidative phosphorylation, oxidative stress (reactive oxygen species), DNA damage, and iron transport, and suggest that lactate dehydrogenase may constitute a useful prognostic and follow-up biomarker. Identifying biomarkers that reflect MPS IVA clinical course, severity, and progression have important implications for disease management. Full article
(This article belongs to the Special Issue New Technologies and Applications in Proteomics Research)
Show Figures

Figure 1

Back to TopTop