Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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 (96)

Search Parameters:
Keywords = mucopolysaccharidoses

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 645 KiB  
Review
Otorhinolaryngological Problems in Mucopolysaccharidoses: A Review of Common Symptoms in a Rare Disease
by Anna Waśniewska-Włodarczyk, Renata Pepaś, Oskar Rosiak and Wiesław Konopka
Brain Sci. 2024, 14(11), 1085; https://doi.org/10.3390/brainsci14111085 - 29 Oct 2024
Viewed by 1120
Abstract
Background: The mucopolysaccharidoses (MPSs) are very rare lysosomal diseases. MPSs belong to inherited diseases; however, newborns are usually asymptomatic. A deficiency of one of the enzymes, which is responsible for glycosaminoglycan (GAG) catabolism, results in the accumulation of this material. GAGs lead to [...] Read more.
Background: The mucopolysaccharidoses (MPSs) are very rare lysosomal diseases. MPSs belong to inherited diseases; however, newborns are usually asymptomatic. A deficiency of one of the enzymes, which is responsible for glycosaminoglycan (GAG) catabolism, results in the accumulation of this material. GAGs lead to progressive damage to tissues. More than 90% of patients with MPS suffer from otitis media with effusion or recurrent otitis media, craniofacial dysmorphia, obstructive sleep apnea, different types of hearing loss, and progressive upper and lower airway dysfunction. Patients visit otolaryngologists often before the diagnosis of MPS. Thus, the awareness of symptoms of MPS is crucial for otolaryngologists and pediatricians. The earlier the diagnosis is made, the more effective treatment is. Ineffective or delayed treatment leads to premature death. Two principal treatments for MPS are currently available: hematopoietic stem cell transplantation (HSCT) and enzyme replacement therapy (ERT). In recent years, there has been a growing interest in gene therapy as a potential treatment for patients with MPS. Mortality in patients with MPS typically occurs during childhood and early adolescence as a consequence of upper and lower respiratory diseases. Methods: This systematic review is based on papers available in the following scientific databases: MEDLINE (via PubMed), Web of Science, Scopus, and the Cochrane Library. Results: After screening, 72 articles met our inclusion criteria. Conclusions: It is of paramount importance that otolaryngologists are involved in this field. This narrative review examines and synthesizes the otolaryngologic issues encountered in patients with MPS. Full article
(This article belongs to the Special Issue Recent Advances in Hearing Impairment)
Show Figures

Figure 1

Figure 1
<p>Craniofacial dysmorphia in patients with MPS. Author: A.W.-W.</p>
Full article ">Figure 2
<p>Otolaryngological problems due to accumulation of GAG.</p>
Full article ">
12 pages, 1120 KiB  
Article
Body Height of MPS I and II Patients after Hematopoietic Stem Cell Transplantation: The Impact of Dermatan Sulphate
by Patryk Lipiński, Agnieszka Różdżyńska-Świątkowska, Agnieszka Ługowska, Jolanta Marucha, Katarzyna Drabko and Anna Tylki-Szymańska
Diagnostics 2024, 14(17), 1956; https://doi.org/10.3390/diagnostics14171956 - 4 Sep 2024
Viewed by 791
Abstract
Introduction: Hematopoietic stem cell transplantation (HSCT) comprises one of the two main treatment regimens for patients with mucopolysaccharidoses (MPS). There is a scarcity of literature concerning the process of growth in children with Mucopolysaccharidosis type I (MPS I) and Mucopolysaccharidosis type I (MPS [...] Read more.
Introduction: Hematopoietic stem cell transplantation (HSCT) comprises one of the two main treatment regimens for patients with mucopolysaccharidoses (MPS). There is a scarcity of literature concerning the process of growth in children with Mucopolysaccharidosis type I (MPS I) and Mucopolysaccharidosis type I (MPS II) after HSCT. The aim of this manuscript was to evaluate the therapeutic effect of HSCT on the heights of patients with MPS I and MPS II. Material and methods: It was an observational, single-center study on patients with MPS I and II treated with HSCT. Results: 6 MPS patients, including 4 MPS I and 2 MPS II, underwent HSCT at a median age of 2 years. All patients are alive to date, with a median age of 7.7 years (range 5.5–12 years) at the last follow-up. In both (MPS I and MPS II) groups of patients treated with HSCT, the growth rate was higher than in untreated patients and was found to be in line with the population norm. In both MPS I and MPS II patients who were treated with HSCT, normalization of urinary GAG excretion was observed. Additionally, no bands of DS and HS in GAG electrophoresis were visible. Conclusions: Both MPS I and MPS II patients presented height gain after HSCT compared to the curves of untreated patients. The absence of dermatan sulphate after HSCT could lead to normal growth in bone length. Full article
(This article belongs to the Special Issue Diagnosis and Management of Metabolic Bone Diseases: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Body height of MPS I patients after HSCT (red lines; arrow—time of HSCT) with comparison with untreated patients (blue line).</p>
Full article ">Figure 2
<p>Body height of MPS II patients after HSCT (red lines; arrow—time of HSCT) with comparison with untreated patients (blue line).</p>
Full article ">Figure 3
<p>Changes in GAG electrophoresis in MPS I patients after HSCT and ERT. Abbreviations: SCh—chondroitin sulphate; SD—dermatan sulphate; SH—heparan sulphate; GAG—glycosaminoglycan; ERT—enzyme replacement therapy; HSCT—hematopoietic stem cell transplantation. (<b>A</b>) 1—before treatment; 2—after 2 years of HSCT; 3—after 4 years of HSCT. (<b>B</b>) 1—before treatment; 2—after 2 years of HSCT; 3—after 3 years of HSCT. (<b>C</b>) 1—before treatment; 2—control sample; 3—after 11 years of ERT; 4—after 21 years of ERT. (<b>D</b>) 1—control sample; 2—before treatment; 3—after 11 years of ERT; 4—after 21 years of ERT.</p>
Full article ">Figure 4
<p>Changes in GAG electrophoresis in MPS II patients after HSCT and ERT. Abbreviations: SCh—chondroitin sulphate; SD—dermatan sulphate; SH—heparan sulphate; GAG—glycosaminoglycan; ERT—enzyme replacement therapy; HSCT—hematopoietic stem cell transplantation. (<b>A</b>) 1—before treatment; 2—after 9 months of HSCT. (<b>B</b>) 1—before treatment; 2—after 3 years of HSCT; 3—after 3 years and 4 months of HSCT. (<b>C</b>) 1—before treatment; 2—after 7.5 years of ERT; 3—after. 2.5 years of ERT; 4—after 16 years of ERT; 5—after 21 years of ERT. (<b>D</b>) 1—before treatment; 2—after 1.5 years of ERT; 3—after 3 years of ERT; 4—after 4.5 years of ERT. (<b>E</b>) 1—before treatment; 2—after 6 years of HSCT; 3—after 10 years of HSCT. (<b>F</b>) 1—MPS II without treatment.</p>
Full article ">
16 pages, 952 KiB  
Review
Mucopolysaccharidosis Type IIIE: A Real Human Disease or a Diagnostic Pitfall?
by Karolina Wiśniewska, Jakub Wolski, Magdalena Żabińska, Aneta Szulc, Lidia Gaffke, Karolina Pierzynowska and Grzegorz Węgrzyn
Diagnostics 2024, 14(16), 1734; https://doi.org/10.3390/diagnostics14161734 - 9 Aug 2024
Cited by 4 | Viewed by 1350
Abstract
Mucopolysaccharidoses (MPS) comprise a group of 12 metabolic disorders where defects in specific enzyme activities lead to the accumulation of glycosaminoglycans (GAGs) within lysosomes. This classification expands to 13 when considering MPS IIIE. This type of MPS, associated with pathogenic variants in the [...] Read more.
Mucopolysaccharidoses (MPS) comprise a group of 12 metabolic disorders where defects in specific enzyme activities lead to the accumulation of glycosaminoglycans (GAGs) within lysosomes. This classification expands to 13 when considering MPS IIIE. This type of MPS, associated with pathogenic variants in the ARSG gene, has thus far been described only in the context of animal models. However, pathogenic variants in this gene also occur in humans, but are linked to a different disorder, Usher syndrome (USH) type IV, which is sparking increasing debate. This paper gathers, discusses, and summarizes arguments both for and against classifying dysfunctions of arylsulfatase G (due to pathogenic variants in the ARSG gene) in humans as another subtype of MPS, called MPS IIIE. Specific difficulties in diagnostics and the classification of some inherited metabolic diseases are also highlighted and discussed. Full article
Show Figures

Figure 1

Figure 1
<p>The pathway of HS degradation with a special emphasis on enzymes for which dysfunctions cause specific subtypes of MPS III. Reactions catalyzed by these enzymes are indicated, while other steps and enzymes for which deficiencies are responsible for other MPS types are shown in a simplified form. Whether deficiency of arylsulfatase G (ARSG) causes MPS IIIE in humans or not is a disputable issue; thus, this MPS subtype is followed by a question mark. Abbreviations: IDS, iduronate-2-sulfatase; IDUA, α-L-iduronidase; ARSG, arylsulfatase G; SGSH, Heparan-<span class="html-italic">N</span>-sulfatase; NAGLU, α-<span class="html-italic">N</span>-acetylglucosaminidase; HGSNAT, heparan α-glucosaminide-<span class="html-italic">N</span>-acetyltransferase; GUSB, β-glucuronidase; GNS, <span class="html-italic">N</span>-acetylglucosamine-6-sulfatase. This scheme was created using BioRender.com (license no. YA2720J3QO).</p>
Full article ">Figure 2
<p>Simplified diagnostic scheme for MPS. Abbreviations: GAG, glycosaminoglycan; LSD, lysosomal storage disease; MPS, mucopolysaccharidosis; MPSPS, mucopolysaccharidosis-plus syndrome.</p>
Full article ">
23 pages, 4212 KiB  
Article
Cellular Organelle-Related Transcriptomic Profile Abnormalities in Neuronopathic Types of Mucopolysaccharidosis: A Comparison with Other Neurodegenerative Diseases
by Karolina Wiśniewska, Lidia Gaffke, Magdalena Żabińska, Grzegorz Węgrzyn and Karolina Pierzynowska
Curr. Issues Mol. Biol. 2024, 46(3), 2678-2700; https://doi.org/10.3390/cimb46030169 - 21 Mar 2024
Cited by 2 | Viewed by 2734
Abstract
Mucopolysaccharidoses (MPS) are a group of diseases caused by mutations in genes encoding lysosomal enzymes that catalyze reactions of glycosaminoglycan (GAG) degradation. As a result, GAGs accumulate in lysosomes, impairing the proper functioning of entire cells and tissues. There are 14 types/subtypes of [...] Read more.
Mucopolysaccharidoses (MPS) are a group of diseases caused by mutations in genes encoding lysosomal enzymes that catalyze reactions of glycosaminoglycan (GAG) degradation. As a result, GAGs accumulate in lysosomes, impairing the proper functioning of entire cells and tissues. There are 14 types/subtypes of MPS, which are differentiated by the kind(s) of accumulated GAG(s) and the type of a non-functional lysosomal enzyme. Some of these types (severe forms of MPS types I and II, MPS III, and MPS VII) are characterized by extensive central nervous system disorders. The aim of this work was to identify, using transcriptomic methods, organelle-related genes whose expression levels are changed in neuronopathic types of MPS compared to healthy cells while remaining unchanged in non-neuronopathic types of MPS. The study was conducted with fibroblast lines derived from patients with neuronopathic and non-neuronopathic types of MPS and control (healthy) fibroblasts. Transcriptomic analysis has identified genes related to cellular organelles whose expression is altered. Then, using fluorescence and electron microscopy, we assessed the morphology of selected structures. Our analyses indicated that the genes whose expression is affected in neuronopathic MPS are often associated with the structures or functions of the cell nucleus, endoplasmic reticulum, or Golgi apparatus. Electron microscopic studies confirmed disruptions in the structures of these organelles. Special attention was paid to up-regulated genes, such as PDIA3 and MFGE8, and down-regulated genes, such as ARL6IP6, ABHD5, PDE4DIP, YIPF5, and CLDN11. Of particular interest is also the GM130 (GOLGA2) gene, which encodes golgin A2, which revealed an increased expression in neuronopathic MPS types. We propose to consider the levels of mRNAs of these genes as candidates for biomarkers of neurodegeneration in MPS. These genes may also become potential targets for therapies under development for neurological disorders associated with MPS and candidates for markers of the effectiveness of these therapies. Although fibroblasts rather than nerve cells were used in this study, it is worth noting that potential genetic markers characteristic solely of neurons would be impractical in testing patients, contrary to somatic cells that can be relatively easily obtained from assessed persons. Full article
(This article belongs to the Special Issue Complex Molecular Mechanism of Monogenic Diseases 2.0)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Number of statistically significant up- and down-regulated transcripts included in organelle-related GO terms, which are Golgi apparatus (GO:0005794); mitochondrion (GO:0005739); ribosome (GO: 0005840); nucleus (GO:0005634); cytoskeleton (GO:0005856); endoplasmic reticulum (GO:0005783); and vacuole (GO:0005773), in neuronopathic MPS types/subtypes relative to control cells (HDFa). Abnormal expression levels of these transcripts were not observed in non-neuronopathic MPS types/subtypes.</p>
Full article ">Figure 2
<p>Heat maps presenting transcripts related to the nucleus (<b>A</b>), endoplasmic reticulum (<b>B</b>), Golgi apparatus (<b>C</b>), mitochondria (<b>D</b>), cytoskeleton (<b>E</b>), lysosome (<b>F</b>), and ribosomes (<b>G</b>) whose expression undergoes particularly high (log<sub>2</sub>FC &gt; 1.5 or log<sub>2</sub>FC &lt; −1.5) changes in neuronopathic MPS types/subtypes relative to control cells (HDFa). Abnormal expression levels of these transcripts were not observed in non-neuronopathic MPS types/subtypes.</p>
Full article ">Figure 3
<p>Morphology of the Golgi apparatus in neuronopathic and non-neuronopathic types/subtypes of MPS relative to control cells (HDFa) as studied by electron microscopy. The analysis of the percentage of fragmented Golgi apparatus structures was performed using at least 100 electron micrographs. Mean values ± SD are presented, with (*) representing statistically significant (<span class="html-italic">p</span> &lt; 0.05) differences relative to the HDFa control and (#) representing statistically significant (<span class="html-italic">p</span> &lt; 0.05) differences between neuronopathic and non-neuronopathic types/subtypes of MPS. Arrows indicate changes in Gogli apparatus morphology.</p>
Full article ">Figure 4
<p>Morphology of the Golgi apparatus in neuronopathic and non-neuronopathic types/subtypes of MPS relative to control cells (HDFa) as studied by fluorescent microscopy using the fluorescent dye CellLight™ Golgi-GFP BacMam 2.0. The analysis of the percentage of fragmented Golgi apparatus structures was performed using at least 30 images. Mean values ± SD are presented, with (*) representing statistically significant (<span class="html-italic">p</span> &lt; 0.05) differences relative to the HDFa control and (#) representing statistically significant (<span class="html-italic">p</span> &lt; 0.05) differences between neuronopathic and non-neuronopathic types/subtypes of MPS.</p>
Full article ">
16 pages, 5195 KiB  
Article
Airway and Anaesthetic Management of Adult Patients with Mucopolysaccharidoses Undergoing Cardiac Surgery
by David Mayhew, Kenneth Palmer, Ian Wilson, Stuart Watson, Karolina M. Stepien, Petra Jenkins and Chaitanya Gadepalli
J. Clin. Med. 2024, 13(5), 1366; https://doi.org/10.3390/jcm13051366 - 28 Feb 2024
Viewed by 1894
Abstract
Background: Mucopolysaccharidoses (MPSs) are rare congenital lysosomal storage disorders due to a deficiency of enzymes metabolising glycosaminoglycans, leading to their accumulation in tissues. This multisystem disease often requires surgical intervention, including valvular cardiac surgery. Adult MPSs have complex airways making anaesthesia risky. Methods: [...] Read more.
Background: Mucopolysaccharidoses (MPSs) are rare congenital lysosomal storage disorders due to a deficiency of enzymes metabolising glycosaminoglycans, leading to their accumulation in tissues. This multisystem disease often requires surgical intervention, including valvular cardiac surgery. Adult MPSs have complex airways making anaesthesia risky. Methods: We report novel three-dimensional (3D) modelling airway assessments and multidisciplinary peri-operative airway management. Results: Five MPS adults underwent cardiac surgery at the national MPS cardiac centre (type I = 4, type II = 1; ages 20, 24, 33, 35, 37 years; two males, three females). All had complex airway abnormalities. Assessments involved examination, nasendoscopy, imaging, functional studies, 3D reconstruction, virtual endoscopy, virtual reality and simulation using computerised, physical modelling. Awake oral fibre-optic intubation was achieved via airway conduit. Staged extubation was performed on the first post-operative day under laryngo-tracheoscopic guidance. The post-operative period involved chest physiotherapy and occupational therapy. All patients had safe intubation, ventilation and extubation. Four had good cardiac surgical outcomes, one (MPS type I; age 35 years) was inoperable due to endocarditis. None had post-operative airway complications. Conclusions: Expertise from cardiovascular-heart team, multidisciplinary airway management, use of novel techniques is vital. Traditional airway assessments are insufficient, so ENT input, radiology and computerised methods to assess and simulate the airway in 3D by collaboration with clinical engineering is essential. Full article
Show Figures

Figure 1

Figure 1
<p>Three-dimensional reconstruction depicting large head, small spine and large jaw in patient two.</p>
Full article ">Figure 2
<p>Acute hyomental angle (39.5 degrees) on the left short hyomental distance (29 mm) in patient three indicating high and anterior larynx.</p>
Full article ">Figure 3
<p>Nasendoscopy view showing large epiglottis, bulky supraglottis narrow supraglottis in patient five.</p>
Full article ">Figure 4
<p>Three-dimensional (3D) reconstruction in patient four showing vertically flattened trachea below sub glottis.</p>
Full article ">Figure 5
<p>Three-dimensional (3D) reconstruction in patient four showing vertically flattened upper trachea, kinking and narrowing of left main bronchus.</p>
Full article ">Figure 6
<p>MADgic<sup>®</sup> device [<a href="#B25-jcm-13-01366" class="html-bibr">25</a>] inside oral cavity, showing that the device bypasses the tongue but the epiglottis sits in the way due to high and anterior larynx.</p>
Full article ">Figure 7
<p>Angulated airway noted on 3D reconstruction.</p>
Full article ">Figure 8
<p>Angulated airway being manipulated utilising virtual reality.</p>
Full article ">Figure 9
<p>Physical three-dimensional printed model of the oropharynx (<b>a</b>), upper airway (<b>b</b>), computer simulation (<b>c</b>) and physical simulation (<b>d</b>).</p>
Full article ">Figure 10
<p>Three-dimensional reconstruction of the chest wall in an adult MPS1 with large spatulate ribs restricting lung expansion. NB: the presence of pre-existing ventriculo-peritoneal shunt.</p>
Full article ">
38 pages, 3780 KiB  
Review
Molecular Mechanisms in Pathophysiology of Mucopolysaccharidosis and Prospects for Innovative Therapy
by Yasuhiko Ago, Estera Rintz, Krishna Sai Musini, Zhengyu Ma and Shunji Tomatsu
Int. J. Mol. Sci. 2024, 25(2), 1113; https://doi.org/10.3390/ijms25021113 - 17 Jan 2024
Cited by 6 | Viewed by 4994
Abstract
Mucopolysaccharidoses (MPSs) are a group of inborn errors of the metabolism caused by a deficiency in the lysosomal enzymes required to break down molecules called glycosaminoglycans (GAGs). These GAGs accumulate over time in various tissues and disrupt multiple biological systems, including catabolism of [...] Read more.
Mucopolysaccharidoses (MPSs) are a group of inborn errors of the metabolism caused by a deficiency in the lysosomal enzymes required to break down molecules called glycosaminoglycans (GAGs). These GAGs accumulate over time in various tissues and disrupt multiple biological systems, including catabolism of other substances, autophagy, and mitochondrial function. These pathological changes ultimately increase oxidative stress and activate innate immunity and inflammation. We have described the pathophysiology of MPS and activated inflammation in this paper, starting with accumulating the primary storage materials, GAGs. At the initial stage of GAG accumulation, affected tissues/cells are reversibly affected but progress irreversibly to: (1) disruption of substrate degradation with pathogenic changes in lysosomal function, (2) cellular dysfunction, secondary/tertiary accumulation (toxins such as GM2 or GM3 ganglioside, etc.), and inflammatory process, and (3) progressive tissue/organ damage and cell death (e.g., skeletal dysplasia, CNS impairment, etc.). For current and future treatment, several potential treatments for MPS that can penetrate the blood–brain barrier and bone have been proposed and/or are in clinical trials, including targeting peptides and molecular Trojan horses such as monoclonal antibodies attached to enzymes via receptor-mediated transport. Gene therapy trials with AAV, ex vivo LV, and Sleeping Beauty transposon system for MPS are proposed and/or underway as innovative therapeutic options. In addition, possible immunomodulatory reagents that can suppress MPS symptoms have been summarized in this review. Full article
Show Figures

Figure 1

Figure 1
<p>Representative inflammatory pathways and activated innate immunity in MPS [<a href="#B226-ijms-25-01113" class="html-bibr">226</a>,<a href="#B227-ijms-25-01113" class="html-bibr">227</a>]. Black arrows indicate activated pathways in MPS. Green arrows indicate pathways that are not considered to be activated. Red arrows indicate enhanced expression of each substance. Short, flat vertical lines at the tips of the dotted lines indicate inhibition by the reagent. Reagents that have been tested in MPS models are colored red. HS: heparan sulfate; TLR: Toll-like receptor; MyD88: myeloid differentiation primary response protein 88; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; ATP: adenosine triphosphate; ROS: reactive oxygen species; TXNIP: thioredoxin-interacting protein; TRX: thioredoxin; NLRP3: NOD-like receptor family pyrin domain-containing 3; IL: interleukin; TNF-α: tumor necrosis factor-alpha; GSDMD: gasdermin D; MMPs: matrix metalloproteinases. This figure was created with <a href="http://Biorender.com" target="_blank">Biorender.com</a> (accessed on 1 December 2023).</p>
Full article ">Figure 2
<p>Receptor-mediated transcytosis across the blood–brain barrier. Receptor-mediated transcytosis (RMT) (molecular Trojan horse) mediates the transfer of enzymes tagged with monoclonal antibodies or ligands targeting specific receptors on the surface of the endothelial cells of the blood–brain barrier. The modality of monoclonal antibodies or ligands targeting transferrin receptors differs depending on the gene product. ArmaGen, Inc. (Calabasas, CA, USA) and JCR Pharmaceuticals Co., Ltd. (Ashiya, Japan)—bivalent (enzyme fused with the whole antibody). DENALI Therapeutics (South San Francisco, CA, USA)—monovalent (enzyme fused with the Fc region of IgG). This figure was created with <a href="http://Biorender.com" target="_blank">Biorender.com</a> (accessed on 1 December 2023).</p>
Full article ">Figure 3
<p>Three-dimensional structural model representing the surface of GALNS (Protein Data Bank ID: 4FDJ), cross-eyed stereo view. The blue area is the surface of the active site consisting of Asp39, Asp40, Arg83, Tyr108, Lys140, His142, His236, Asp288, Asn289, Lys310, and dihydroxyalanine 79. The red area is the surface of Trp520. These two areas are in contact. This model does not determine the location of two amino acid residues, Ser 521 and His 522, at the C-terminus. The molecule depicted in the stick model is N-acetylgalactosamine. This figure was created with PyMOL Version 2.5.2.</p>
Full article ">Figure 4
<p>Electrostatic potential of AAV capsid protein and its affinity to targets. (<b>A</b>) Heparan sulfate proteoglycan (HSPG) is negatively charged with disaccharide units of β-D-glucuronic acid (GlcA) and N-acetylglucosamine (GlcNAc). It has an affinity towards the positively charged HSPG-binding domain of AAV capsid, which consists of basic amino acids such as Arg and Lys. (<b>B</b>) Modification of VP1 and VP2 capsid proteins with negatively charged amino acid (Asp) on AAV increases the affinity of the modified vector for positively charged calcium hydroxyapatite in bone matrix. This figure was created with <a href="http://Biorender.com" target="_blank">Biorender.com</a> (accessed on 1 December 2023).</p>
Full article ">
7 pages, 680 KiB  
Case Report
Extensive and Persistent Dermal Melanocytosis in a Male Carrier of Mucopolysaccharidosis Type IIIC (Sanfilippo Syndrome): A Case Report
by Maurizio Romagnuolo, Chiara Moltrasio, Serena Gasperini, Angelo Valerio Marzano and Stefano Cambiaghi
Children 2023, 10(12), 1920; https://doi.org/10.3390/children10121920 - 13 Dec 2023
Cited by 2 | Viewed by 2018
Abstract
Congenital dermal melanocytosis (DM) represents a common birthmark mainly found in children of Asian and darker skin phototype descent, clinically characterized by an oval blue-grey macule or macules, commonly located on the lumbosacral area. In rare DM cases, when presenting with diffuse macules [...] Read more.
Congenital dermal melanocytosis (DM) represents a common birthmark mainly found in children of Asian and darker skin phototype descent, clinically characterized by an oval blue-grey macule or macules, commonly located on the lumbosacral area. In rare DM cases, when presenting with diffuse macules persisting during the first years of life, it could represent a cutaneous feature of mucopolysaccharidoses (MPS). Extensive congenital DM is actually associated with Hurler syndrome (MPS type I) and Hunter syndrome (MPS type II), although several reports also described this association with MPS type VI and other lysosomal storage disorders (LySD), including GM1 gangliosidosis, mucolipidosis, Sandhoff disease, and Niemann–Pick disease. Here, we present the case of a two-year-old boy presenting with extensive dermal melanocytosis, generalized hypertrichosis, and chronic itch, harboring a heterozygous variant of uncertain significance, NM_152419.3: c.493C>T (p.Pro165Ser), in the exon 4 of HGSNAT gene, whose mutations are classically associated with MPS IIIC, also known as Sanfilippo syndrome. This is the first report that highlights the association between extensive congenital DM and MPS type IIIC, as well as a pathogenetic link between heterozygous LySD carrier status and congenital DM. We speculate that some cases of extensive congenital DM could be related to heterozygous LySD carriers, as a manifestation of a mild clinical phenotype. Full article
(This article belongs to the Special Issue Reviews in Pediatric Dermatology)
Show Figures

Figure 1

Figure 1
<p>Clinical photographs of the 4-year-old boy showing (<b>a</b>) an irregularly shaped light brown patch on the right arm consistent with a jagged cafè-au-lait spot; (<b>b</b>) an hyperpigmented (light brown) mosaic along the left latero-cervical region with an irregular lateral border and a sharp edge on the midline; (<b>c</b>) diffuse dermal melanocytosis, presenting as multiple blue-grey oval-shaped patches on the back of the patient; and (<b>d</b>) a detail of a dermal melanocytosis patch. Generalized hypertrichosis could be appreciated in all photographs.</p>
Full article ">
12 pages, 1595 KiB  
Article
New Perspectives in Dried Blood Spot Biomarkers for Lysosomal Storage Diseases
by Justyna Spiewak, Ivan Doykov, Apostolos Papandreou, Jenny Hällqvist, Philippa Mills, Peter T. Clayton, Paul Gissen, Kevin Mills and Wendy E. Heywood
Int. J. Mol. Sci. 2023, 24(12), 10177; https://doi.org/10.3390/ijms241210177 - 15 Jun 2023
Cited by 8 | Viewed by 2646
Abstract
Dried blood spots (DBSs) biomarkers are convenient for monitoring for specific lysosomal storage diseases (LSDs), but they could have relevance for other LSDs. To determine the specificity and utility of glycosphingolipidoses biomarkers against other LSDs, we applied a multiplexed lipid liquid chromatography tandem [...] Read more.
Dried blood spots (DBSs) biomarkers are convenient for monitoring for specific lysosomal storage diseases (LSDs), but they could have relevance for other LSDs. To determine the specificity and utility of glycosphingolipidoses biomarkers against other LSDs, we applied a multiplexed lipid liquid chromatography tandem mass spectrometry assay to a DBS cohort of healthy controls (n = 10) and Gaucher (n = 4), Fabry (n = 10), Pompe (n = 2), mucopolysaccharidosis types I–VI (n = 52), and Niemann–Pick disease type C (NPC) (n = 5) patients. We observed no complete disease specificity for any of the markers tested. However, comparison among the different LSDs highlighted new applications and perspectives of the existing biomarkers. We observed elevations in glucosylceramide isoforms in the NPC and Gaucher patients relative to the controls. In NPC, there was a greater proportion of C24 isoforms, giving a specificity of 96–97% for NPC, higher than 92% for the NPC biomarker N-palmitoyl-O-phosphocholineserine ratio to lyso-sphingomyelin. We also observed significantly elevated levels of lyso-dihexosylceramide in Gaucher and Fabry disease as well as elevated lyso-globotriaosylceramide (Lyso-Gb3) in Gaucher disease and the neuronopathic forms of Mucopolysaccharidoses. In conclusion, DBS glucosylceramide isoform profiling has increased the specificity for the detection of NPC, thereby improving diagnostic accuracy. Low levels of lyso-lipids can be observed in other LSDs, which may have implications in their disease pathogenesis. Full article
(This article belongs to the Special Issue Biomarkers in Rare Diseases 3.0)
Show Figures

Figure 1

Figure 1
<p>The glycosphingolipid degradation pathway, illustrating known enzyme defects and associated disorders (red text). Lipids that are included in the assay are indicated by *. PPCS presented individually, as the associated pathway is unknown.</p>
Full article ">Figure 2
<p>Dried blood spot glycosphingolipid levels in a cohort of patients with lysosomal storage diseases. The four main GSL species detectable in DBS are shown. (<b>A</b>) GlcCer—only significantly different to the controls for NPC, and therefore useful as a biomarker. (<b>B</b>) CDH—no significant changes observed. (<b>C</b>) Gb3 CDH—no significant changes observed. (<b>D</b>) GB4 CDH—no significant changes observed. Patients in treatment are represented by half shaded circles. Gaucher type I is represented by a triangle symbol, and type III, by a circle. The untreated MPS type I group is depicted as follows: Hurler in orange, Hurler–Scheie in grey, and Scheie in pink. Significance determined by Kruskal–Wallis non-parametric test. * means significance of <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 3
<p>Glucosylceramide isoforms in dried blood spots from patients with lysosomal storage diseases. (<b>A</b>) A summary pie chart of the mean values of each isoform in the control, Gaucher, and NPC disease, showing a differing isoform profile in NPC disease. (<b>B</b>) The C16 isoform in Gaucher and NPC DBS in comparison with control levels. (<b>C</b>) The C24:1 isoform in Gaucher and NPC DBS in comparison with control levels. (<b>D</b>) Ratio of GlcCer C24:1/C16 isoforms in a cohort of patients with LSDs, showing that NPC has the greatest ratio difference relative to controls. (<b>E</b>) Ratio of GlcCer 24:2-OH/CDH 24:2-OH isoforms, showing the best specificity for NPC. Patients on treatment are represented by half-shaded circles. Gaucher type I is indicated by a triangle symbol, and type III, by a circle. The untreated MPS type I group circles are coloured as follows: Hurler as orange, Hurler–Scheie as grey, and Scheie as pink. **** means significance of <span class="html-italic">p</span> ≤ 0.0001; ** means significance of <span class="html-italic">p</span> ≤ 0.01; * means significance of <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 4
<p>Lyso-glycosphingolipid dried-blood-spot analysis of samples from a cohort of patients with lysosomal storage diseases. (<b>A</b>) The Gaucher disease group has the highest levels of lyso-Gb1. Low-level changes in lyso-Gb1 in other LSDs were observed. (<b>B</b>) Small increases in the levels of lyso-CDH were seen in the Fabry and Gaucher samples. (<b>C</b>) The Fabry disease group has the highest lyso-Gb3 levels. Much smaller changes were observed for the other LSDs patients on treatment, represented by half-shaded circles. Gaucher type I samples are represented by a triangle symbol, and type III, by a circle. The untreated MPS type I group circles are coloured as follows: Hurler is orange, Hurler–Scheie is grey, and Scheie is pink. **** means significance of <span class="html-italic">p</span> ≤ 0.0001; ** means significance of <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 5
<p>Levels of NPC biomarkers in a cohort of patients with lysosomal storage diseases. (<b>A</b>) Levels of total sphingomyelin in the LSD groups are similar to those seen in control samples. (<b>B</b>) Lyso-sphingomyelin levels are similar to those seen in the control cohort, with some outlier patients. (<b>C</b>) Total PPCS is significantly elevated in NPC but shows outliers in other disorders. (<b>D</b>) Ratioing of PPCS to lyso-sphingomyelin improves specificity for NPC relative to other LSDs. Patients on treatment are represented by half-shaded circles. Gaucher type I is indicated by a triangle symbol, and type III, by a circle. The untreated MPS type I group circles are coloured as follows: Hurler in orange, Hurler–Scheie in grey, and Scheie in pink. *** significance of <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">
14 pages, 2015 KiB  
Article
Leukocyte Imbalances in Mucopolysaccharidoses Patients
by Nuno Lopes, Maria L. Maia, Cátia S. Pereira, Inês Mondragão-Rodrigues, Esmeralda Martins, Rosa Ribeiro, Ana Gaspar, Patrício Aguiar, Paula Garcia, Maria Teresa Cardoso, Esmeralda Rodrigues, Elisa Leão-Teles, Roberto Giugliani, Maria F. Coutinho, Sandra Alves and M. Fátima Macedo
Biomedicines 2023, 11(6), 1699; https://doi.org/10.3390/biomedicines11061699 - 13 Jun 2023
Cited by 2 | Viewed by 1841
Abstract
Mucopolysaccharidoses (MPSs) are rare inherited lysosomal storage diseases (LSDs) caused by deficient activity in one of the enzymes responsible for glycosaminoglycans lysosomal degradation. MPS II is caused by pathogenic mutations in the IDS gene, leading to deficient activity of the enzyme iduronate-2-sulfatase, which [...] Read more.
Mucopolysaccharidoses (MPSs) are rare inherited lysosomal storage diseases (LSDs) caused by deficient activity in one of the enzymes responsible for glycosaminoglycans lysosomal degradation. MPS II is caused by pathogenic mutations in the IDS gene, leading to deficient activity of the enzyme iduronate-2-sulfatase, which causes dermatan and heparan sulfate storage in the lysosomes. In MPS VI, there is dermatan sulfate lysosomal accumulation due to pathogenic mutations in the ARSB gene, leading to arylsulfatase B deficiency. Alterations in the immune system of MPS mouse models have already been described, but data concerning MPSs patients is still scarce. Herein, we study different leukocyte populations in MPS II and VI disease patients. MPS VI, but not MPS II patients, have a decrease percentage of natural killer (NK) cells and monocytes when compared with controls. No alterations were identified in the percentage of T, invariant NKT, and B cells in both groups of MPS disease patients. However, we discovered alterations in the naïve versus memory status of both helper and cytotoxic T cells in MPS VI disease patients compared to control group. Indeed, MPS VI disease patients have a higher frequency of naïve T cells and, consequently, lower memory T cell frequency than control subjects. Altogether, these results reveal MPS VI disease-specific alterations in some leukocyte populations, suggesting that the type of substrate accumulated and/or enzyme deficiency in the lysosome may have a particular effect on the normal cellular composition of the immune system. Full article
(This article belongs to the Special Issue Inherited Metabolic Disorders: From Bench to Bedside)
Show Figures

Figure 1

Figure 1
<p>Major peripheral blood mononuclear cells (PBMCs) populations in MPS II, MPS VI disease patients, and control subjects. (<b>A</b>) Gating strategy used to identify T cells (CD3<sup>+</sup>); B cells (CD19<sup>+</sup>); NK cells (CD56<sup>+</sup> CD3<sup>−</sup>) and monocytes (CD14<sup>+</sup>) populations. (<b>B</b>) Percentage of T cells, B cells and NK cells was determined among lymphocytes and of monocytes among PBMCs. Horizontal bars represent mean values. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Frequency of naïve, central memory, effector memory CD4<sup>+</sup> and CD8<sup>+</sup> T cell subsets in MPS II, MPS VI disease patients, and control subjects. (<b>A</b>) Gating strategy applied selecting within the lymphocyte gate the populations of CD4<sup>+</sup>CD3<sup>+</sup> (T helper cells) and CD8<sup>+</sup>CD3<sup>+</sup> (cytotoxic T cells). Within each population: naïve, central memory (CM) and effector memory (EM) cell subset were detailed. (<b>B</b>) Percentage of CD4<sup>+</sup> T cells in total lymphocytes, (<b>C</b>) Percentage of CD8<sup>+</sup> T cells in total lymphocytes, (<b>D</b>) Percentage of naïve cells in CD4<sup>+</sup> T cells, (<b>E</b>) Percentage of CM cells in CD4<sup>+</sup> T cells, (<b>F</b>) Percentage of EM cells in CD4<sup>+</sup> T cells, (<b>G</b>) Percentage of naïve cells in CD8<sup>+</sup> T cells, (<b>H</b>) Percentage of CM cells in CD8<sup>+</sup> T cells, and (<b>I</b>) Percentage of EM cells in CD8<sup>+</sup> T cells. Horizontal bars represent mean values. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Percentage and phenotype of iNKT cells in MPS disease patients, and control subjects. (<b>A</b>) Gating strategy to characterize iNKT cells by the expression of CD4, CD8 and CD161 among total T cells. (<b>B</b>) Percentage of iNKT cells in total T cells of MPS II disease patients and controls, (<b>C</b>) CD4<sup>+</sup>CD8<sup>−</sup> cells in iNKT cells of MPS II disease patients and controls, (<b>D</b>) CD4<sup>−</sup>CD8<sup>+</sup> cells in iNKT cells of MPS II disease patients and controls, (<b>E</b>) CD8<sup>−</sup>/double negative (DN) cells in iNKT cells of MPS II disease patients and controls and (<b>F</b>) CD161<sup>+</sup> cells in iNKT cells of MPS II and VI disease patients and control group. Horizontal bars represent mean values. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
29 pages, 4247 KiB  
Review
Neurological Disease Modeling Using Pluripotent and Multipotent Stem Cells: A Key Step towards Understanding and Treating Mucopolysaccharidoses
by Sofia Carvalho, Juliana Inês Santos, Luciana Moreira, Mariana Gonçalves, Hugo David, Liliana Matos, Marisa Encarnação, Sandra Alves and Maria Francisca Coutinho
Biomedicines 2023, 11(4), 1234; https://doi.org/10.3390/biomedicines11041234 - 21 Apr 2023
Cited by 2 | Viewed by 3519
Abstract
Despite extensive research, the links between the accumulation of glycosaminoglycans (GAGs) and the clinical features seen in patients suffering from various forms of mucopolysaccharidoses (MPSs) have yet to be further elucidated. This is particularly true for the neuropathology of these disorders; the neurological [...] Read more.
Despite extensive research, the links between the accumulation of glycosaminoglycans (GAGs) and the clinical features seen in patients suffering from various forms of mucopolysaccharidoses (MPSs) have yet to be further elucidated. This is particularly true for the neuropathology of these disorders; the neurological symptoms are currently incurable, even in the cases where a disease-specific therapeutic approach does exist. One of the best ways to get insights on the molecular mechanisms driving that pathogenesis is the analysis of patient-derived cells. Yet, not every patient-derived cell recapitulates relevant disease features. For the neuronopathic forms of MPSs, for example, this is particularly evident because of the obvious inability to access live neurons. This scenario changed significantly with the advent of induced pluripotent stem cell (iPSC) technologies. From then on, a series of differentiation protocols to generate neurons from iPSC was developed and extensively used for disease modeling. Currently, human iPSC and iPSC-derived cell models have been generated for several MPSs and numerous lessons were learnt from their analysis. Here we review most of those studies, not only listing the currently available MPS iPSC lines and their derived models, but also summarizing how they were generated and the major information different groups have gathered from their analyses. Finally, and taking into account that iPSC generation is a laborious/expensive protocol that holds significant limitations, we also hypothesize on a tempting alternative to establish MPS patient-derived neuronal cells in a much more expedite way, by taking advantage of the existence of a population of multipotent stem cells in human dental pulp to establish mixed neuronal and glial cultures. Full article
Show Figures

Figure 1

Figure 1
<p>The four major types of studies involving the generation and characterization of induced pluripotent stem cells (iPSCs) as in vitro models for Mucopolysaccharidoses (MPSs), grouped together according to their ultimate aims: (1) papers on iPSC generation and characterization; (2) papers describing their subsequent differentiation into different types of neuronal cells; (3) papers on the use of iPSCs and their derived cell models for drug screening; and (4) papers on the use of genetically corrected iPSCs for therapeutic purposes.</p>
Full article ">Figure 2
<p>Advantages and limitations of iPSCs. Even though extensively reported in the literature, the issues marked with (*) have been largely solved thanks to the most recent technical improvements of the iPSC generation protocols.</p>
Full article ">Figure 3
<p>Different sources of mesenchymal stem cells (MSCs).</p>
Full article ">Figure 4
<p>Minimal requirements for identification of mesenchymal stem cells (MSCs).</p>
Full article ">Figure 5
<p>Schematic drawing illustrating the sources of dental mesenchymal stem cells (DMSCs) in the oral cavity. Abbreviations: GMSCs, gingiva-derived MSCs; DFPCs, dental follicle progenitor cells; SHED, stem cells from exfoliated deciduous teeth; SCAPs, stem cells from the apical papilla; DPSCs, dental pulp stem cells; PDLSCs, periodontal ligament stem cells.</p>
Full article ">Figure 6
<p>Advantages and limitations of dental pulp stem cells (DPSCs).</p>
Full article ">
34 pages, 1855 KiB  
Review
Redox Imbalance in Neurological Disorders in Adults and Children
by Federica Rey, Clarissa Berardo, Erika Maghraby, Alessia Mauri, Letizia Messa, Letizia Esposito, Giovanna Casili, Sara Ottolenghi, Eleonora Bonaventura, Salvatore Cuzzocrea, Gianvincenzo Zuccotti, Davide Tonduti, Emanuela Esposito, Irene Paterniti, Cristina Cereda and Stephana Carelli
Antioxidants 2023, 12(4), 965; https://doi.org/10.3390/antiox12040965 - 20 Apr 2023
Cited by 7 | Viewed by 4983
Abstract
Oxygen is a central molecule for numerous metabolic and cytophysiological processes, and, indeed, its imbalance can lead to numerous pathological consequences. In the human body, the brain is an aerobic organ and for this reason, it is very sensitive to oxygen equilibrium. The [...] Read more.
Oxygen is a central molecule for numerous metabolic and cytophysiological processes, and, indeed, its imbalance can lead to numerous pathological consequences. In the human body, the brain is an aerobic organ and for this reason, it is very sensitive to oxygen equilibrium. The consequences of oxygen imbalance are especially devastating when occurring in this organ. Indeed, oxygen imbalance can lead to hypoxia, hyperoxia, protein misfolding, mitochondria dysfunction, alterations in heme metabolism and neuroinflammation. Consequently, these dysfunctions can cause numerous neurological alterations, both in the pediatric life and in the adult ages. These disorders share numerous common pathways, most of which are consequent to redox imbalance. In this review, we will focus on the dysfunctions present in neurodegenerative disorders (specifically Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis) and pediatric neurological disorders (X-adrenoleukodystrophies, spinal muscular atrophy, mucopolysaccharidoses and Pelizaeus–Merzbacher Disease), highlighting their underlining dysfunction in redox and identifying potential therapeutic strategies. Full article
(This article belongs to the Special Issue Redox Signaling Regulation in Neurological Disorders)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the alterations in redox mechanisms that can lead to numerous aberrant processes, and ultimately lead to neurological disorders.</p>
Full article ">Figure 2
<p>Schematic representation of mitochondrial damage mechanisms in some examples of pediatric-onset neurodegenerative diseases.</p>
Full article ">
8 pages, 1108 KiB  
Communication
Positive Impact of Home ERT for Mucopolysaccharidoses and Pompe Disease: The Lesson Learnt from the COVID-19 Pandemic
by Agata Fiumara, Giuseppina Lanzafame, Annamaria Sapuppo, Alessia Arena, Lara Cirnigliaro and Rita Barone
Healthcare 2023, 11(8), 1176; https://doi.org/10.3390/healthcare11081176 - 19 Apr 2023
Cited by 2 | Viewed by 1836
Abstract
Objective: Patients with Lysosomal disorders (LSDs) are treated with regular infusions of enzyme replacement therapy (ERT). During the COVID-19 pandemic, home treatment was permitted. This study aimed at monitoring the patients’ compliance with home therapy and its effects on physical, psychological, and relational [...] Read more.
Objective: Patients with Lysosomal disorders (LSDs) are treated with regular infusions of enzyme replacement therapy (ERT). During the COVID-19 pandemic, home treatment was permitted. This study aimed at monitoring the patients’ compliance with home therapy and its effects on physical, psychological, and relational issues. Moreover, we also tested the possible impact of home therapy on familial relationships and contacts with the referral hospital. Materials and Methods: Thirteen patients with Pompe disease (N = 8) and MPS (N = 5) were tested through an online questionnaire designed to assess their level of appreciation and satisfaction with home therapy and their feelings about the referral centre and psychological support provided. Results: Most of the patients (84%) stressed the positive impact of home therapy. All patients described a significant reduction in stressful conditions associated with the need to attend the hospital every week or two. Conclusions: Home ERT leads to a clear improvement in “daily life skills”, as represented in our by sample by positive feelings, better emotional self-control, and an increased ability to understand the feelings of relatives. Our data underline the paramount positive effect home ERT has on both patients and their families. Full article
Show Figures

Figure 1

Figure 1
<p>Reported changes related to home therapy in the study patients.</p>
Full article ">Figure 2
<p>(<b>A</b>) Appreciation of psychological support. The majority of our patients were completely (46%) or quite satisfied (39%) with the psychological support and guidance offered both pre- and post-transition to their homes (<b>B</b>) Type of changes due to psychological support.</p>
Full article ">Figure 3
<p>Perceived emotions related to a new lockdown risk.</p>
Full article ">
10 pages, 1957 KiB  
Article
Mucopolysaccharidoses Differential Diagnosis by Mass Spectrometry-Based Analysis of Urine Free Glycosaminoglycans—A Diagnostic Prediction Model
by Francesca D’Avanzo, Alessandra Zanetti, Andrea Dardis, Maurizio Scarpa, Nicola Volpi, Francesco Gatto and Rosella Tomanin
Biomolecules 2023, 13(3), 532; https://doi.org/10.3390/biom13030532 - 15 Mar 2023
Cited by 2 | Viewed by 1844
Abstract
Impaired glycosaminoglycans (GAGs) catabolism may lead to a cluster of rare metabolic and genetic disorders called mucopolysaccharidoses (MPSs). Each subtype is caused by the deficiency of one of the lysosomal hydrolases normally degrading GAGs. Affected tissues accumulate undegraded GAGs in cell lysosomes and [...] Read more.
Impaired glycosaminoglycans (GAGs) catabolism may lead to a cluster of rare metabolic and genetic disorders called mucopolysaccharidoses (MPSs). Each subtype is caused by the deficiency of one of the lysosomal hydrolases normally degrading GAGs. Affected tissues accumulate undegraded GAGs in cell lysosomes and in the extracellular matrix, thus leading to the MPS complex clinical phenotype. Although each MPS may present with recognizable signs and symptoms, these may often overlap between subtypes, rendering the diagnosis difficult and delayed. Here, we performed an exploratory analysis to develop a model that predicts MPS subtypes based on UHPLC-MS/MS measurement of a urine free GAG profile (or GAGome). We analyzed the GAGome of 78 subjects (38 MPS, 37 healthy and 3 with other MPS symptom-overlapping disorders) using a standardized kit in a central-blinded laboratory. We observed several MPS subtype-specific GAGome changes. We developed a multivariable penalized Lasso logistic regression model that attained 91.2% balanced accuracy to distinguish MPS type II vs. III vs. any other subtype vs. not MPS, with sensitivity and specificity ranging from 73.3% to 91.7% and from 98.4% to 100%, depending on the predicted subtype. In conclusion, the urine GAGome was revealed to be useful in accurately discriminating the different MPS subtypes with a single UHPLC-MS/MS run and could serve as a reliable diagnostic test for a more rapid MPS biochemical diagnosis. Full article
Show Figures

Figure 1

Figure 1
<p>Total CS, HA and HS concentrations (µg/mg creatinine) measured in the different groups of MPS patients and controls (non-MPS patients—ML II, SMDK—as well as healthy subjects). SMDK: spondylometaphyseal dysplasia-Kozlowski type. ML II: mucolipidosis type II.</p>
Full article ">Figure 2
<p>Relative concentration (in mass fraction %) of CS (upper panel) and HS (lower panel) disaccharides in the groups. SMDK: spondylometaphyseal dysplasia-Kozlowski type. ML II: mucolipidosis type II. See also <a href="#app1-biomolecules-13-00532" class="html-app">Table S2</a>.</p>
Full article ">Figure 3
<p>Heatmap of the GAGome profile across MPS subtypes compared to the control group. The color map is indicative of the standardized mean change for each GAGome measurement compared to the control group (red = increase and blue = decrease). Stars indicate statistically significant differences vs. the control group.</p>
Full article ">Figure 4
<p>Prediction probability (in %) for each class as obtained by the prediction model developed for differential diagnosis of MPS subtypes, given the urine free GAGome measurements as inputs, vs. the observed diagnosis. SMDK: spondylometaphyseal dysplasia-Kozlowski type. ML II: mucolipidosis type II.</p>
Full article ">
12 pages, 882 KiB  
Article
Individual Treatment Trials—Do Experts Know and Use This Option to Improve the Treatability of Mucopolysaccharidosis?
by Anna-Maria Wiesinger, Hannah Strobl and Florian B. Lagler
Pharmaceuticals 2023, 16(3), 416; https://doi.org/10.3390/ph16030416 - 9 Mar 2023
Cited by 4 | Viewed by 2292
Abstract
Mucopolysaccharidoses (MPS) are a group of rare, heterogeneous, lysosomal storage disorders. Patients show a broad spectrum of clinical features with a substantial unmet medical need. Individual treatment trials (ITTs) might be a valid, time- and cost-efficient way to facilitate personalized medicine in the [...] Read more.
Mucopolysaccharidoses (MPS) are a group of rare, heterogeneous, lysosomal storage disorders. Patients show a broad spectrum of clinical features with a substantial unmet medical need. Individual treatment trials (ITTs) might be a valid, time- and cost-efficient way to facilitate personalized medicine in the sense of drug repurposing in MPS. However, this treatment option has so far hardly been used—at least hardly been reported or published. Therefore, we aimed to investigate the awareness and utilization of ITTs among MPS clinicians, as well as the potential challenges and innovative approaches to overcome key hurdles, by using an international expert survey on ITTs, namely, ESITT. Although 74% (20/27) were familiar with the concept of ITTs, only 37% (10/27) ever used it, and subsequently only 15% (2/16) published their results. The indicated hurdles of ITTs in MPS were mainly the lack of time and know-how. An evidence-based tool, which provides resources and expertise needed for high-quality ITTs, was highly appreciated by the vast majority (89%; 23/26). The ESITT highlights a serious deficiency of ITT implementation in MPS—a promising option to improve its treatability. Furthermore, we discuss the challenges and innovative approaches to overcome key barriers to ITTs in MPS. Full article
(This article belongs to the Special Issue Development of Medicines for Rare Pediatric Diseases)
Show Figures

Figure 1

Figure 1
<p>Response to the survey question “Have you ever heard of n-of-1-trials?” (left pie chart) and response to the survey question “Have you ever used n-of-1 trials in the treatment of your MPS patients?” (right pie chart); <span class="html-italic">n</span> = 27.</p>
Full article ">Figure 2
<p>Flow chart overview of all participating MPS clinicians with regard to previous know-how and implementation of ITTs. Out of the 27 experts, 20 had already heard about ITTs and 14 had conducted ITTs—either with MPS patients or MPS and another disorder or only other disorders. A “single expert” is defined as a standalone expert, while “&gt;1 expert” means a kind of interdisciplinary expert team.</p>
Full article ">Figure 3
<p>Response to the survey question “Assume there is a service that made it easy for you to offer n-of-1 trials to select patients in your practice with MPS. How likely are you to use a service like this to make data driven treatment choices at least once in the next year?” (<span class="html-italic">n</span> = 26).</p>
Full article ">
18 pages, 3878 KiB  
Article
Decreased Levels of Chaperones in Mucopolysaccharidoses and Their Elevation as a Putative Auxiliary Therapeutic Approach
by Magdalena Żabińska, Lidia Gaffke, Patrycja Bielańska, Magdalena Podlacha, Estera Rintz, Zuzanna Cyske, Grzegorz Węgrzyn and Karolina Pierzynowska
Pharmaceutics 2023, 15(2), 704; https://doi.org/10.3390/pharmaceutics15020704 - 20 Feb 2023
Cited by 4 | Viewed by 3132
Abstract
Mucopolysaccharidoses (MPS) are rare genetic disorders belonging to the lysosomal storage diseases. They are caused by mutations in genes encoding lysosomal enzymes responsible for degrading glycosaminoglycans (GAGs). As a result, GAGs accumulate in lysosomes, leading to impairment of cells, organs and, consequently, the [...] Read more.
Mucopolysaccharidoses (MPS) are rare genetic disorders belonging to the lysosomal storage diseases. They are caused by mutations in genes encoding lysosomal enzymes responsible for degrading glycosaminoglycans (GAGs). As a result, GAGs accumulate in lysosomes, leading to impairment of cells, organs and, consequently, the entire body. Many of the therapies proposed thus far require the participation of chaperone proteins, regardless of whether they are therapies in common use (enzyme replacement therapy) or remain in the experimental phase (gene therapy, STOP-codon-readthrough therapy). Chaperones, which include heat shock proteins, are responsible for the correct folding of other proteins to the most energetically favorable conformation. Without their appropriate levels and activities, the correct folding of the lysosomal enzyme, whether supplied from outside or synthesized in the cell, would be impossible. However, the baseline level of nonspecific chaperone proteins in MPS has never been studied. Therefore, the purpose of this work was to determine the basal levels of nonspecific chaperone proteins of the Hsp family in MPS cells and to study the effect of normalizing GAG concentrations on these levels. Results of experiments with fibroblasts taken from patients with MPS types I, II, IIIA, IIIB, IIIC, IID, IVA, IVB, VI, VII, and IX, as well as from the brains of MPS I mice (Idua−/−), indicated significantly reduced levels of the two chaperones, Hsp70 and Hsp40. Interestingly, the reduction in GAG levels in the aforementioned cells did not lead to normalization of the levels of these chaperones but caused only a slight increase in the levels of Hsp40. An additional transcriptomic analysis of MPS cells indicated that the expression of other genes involved in protein folding processes and the cell response to endoplasmic reticulum stress, resulting from the appearance of abnormally folded proteins, was also modulated. To summarize, reduced levels of chaperones may be an additional cause of the low activity or inactivity of lysosomal enzymes in MPS. Moreover, this may point to causes of treatment failure where the correct structure of the enzyme supplied or synthesized in the cell is crucial to lower GAG levels. Full article
(This article belongs to the Special Issue Novel Therapeutic Approaches in Rare Genetic Diseases)
Show Figures

Figure 1

Figure 1
<p>Decreased levels of Hsp70 in MPS cells and effects of ERT and SRT. Western blotting (<b>A</b>,<b>B</b>) and immunofluorescence (<b>C</b>,<b>D</b>) analyses of the abundance of Hsp70 in MPS I and MPS II fibroblasts, relative to HDFa control cells, either non-treated (minuses) or treated (pluses) with Aldurazyme (ALD, recombinant α-L-iduronidase) at 0.58 mg/mL, Elaprase (ELA, recombinant 2-iduronate sulfatase) at 0.5 mg/mL, DMSO (solvent for genistein) at 0.05%, or genistein (GEN) at 50 μM for 24 h. Panel (<b>A</b>) shows a representative Western blot (with GAPDH as loading control) while panel (<b>B</b>) demonstrates quantitative analyses (based on densitometry) from 3 independent experiments, with error bars representing SD. Panel (<b>C</b>) shows representative fluorescent microscopic pictures (with scale bars indicating 50 μm), while panel (<b>D</b>) demonstrates quantitative analyses (relative fluorescence intensity) from 100 randomly chosen cells of each variant of the experiment. In panels (<b>B</b>) and (<b>D</b>), asterisks indicate statistically significant differences (at <span class="html-italic">p</span> &lt; 0.05 in two-way ANOVA and post hoc Tukey’s test) relative to HDFa cells. No statistically significant differences were detected between non-treated and treated MPS I or MPS II fibroblasts.</p>
Full article ">Figure 2
<p>Decreased levels of Hsp40 in MPS cells and effects of ERT and SRT. Western blotting (<b>A</b>,<b>B</b>) and immunofluorescence (<b>C</b>,<b>D</b>) analyses of the abundance of Hsp40 in MPS I and MPS II fibroblasts, relative to HDFa control cells, either non-treated (minuses) or treated (pluses) with Aldurazyme (ALD, recombinant α-L-iduronidase) at 0.58 mg/mL, Elaprase (ELA, recombinant 2-iduronate sulfatase) at 0.5 mg/mL, DMSO (solvent for genistein) at 0.05%, or genistein (GEN) at 50 μM for 24 h. Panel (<b>A</b>) shows a representative Western-blot (with GAPDH as loading control) while panel (<b>B</b>) demonstrates quantitative analyses (based on densitometry) from 3 independent experiments with error bars representing SD. Panel (<b>C</b>) shows representative fluorescent microscopic pictures (with scale bars indicating 25 μm), while panel (<b>D</b>) demonstrates quantitative analyses (relative fluorescence intensity) from 100 randomly chosen cells of each variant of the experiment. In panels (<b>B</b>) and (<b>D</b>), asterisks indicate statistically significant differences (at <span class="html-italic">p</span> &lt; 0.05) relative to HDFa cells, and hashtags indicate statistically significant differences (at <span class="html-italic">p</span> &lt; 0.05 in two-way ANOVA and post hoc Tukey’s test) relative to non-treated MPS I or MPS II fibroblasts.</p>
Full article ">Figure 3
<p>Elevated GAG levels in MPS cells and storage reduction by ERT or SRT. GAG levels in MPS I and MPS II fibroblasts, relative to HDFa control cells, either non-treated or treated with Aldurazyme (ALD, recombinant α-L-iduronidase) at 0.58 mg/mL, Elaprase (ELA, recombinant 2-iduronate sulfatase) at 0.5 mg/mL, DMSO (solvent for genistein) at 0.05%, or genistein (GEN) at 50 μM for 24 h. Asterisks indicate statistically significant differences (at <span class="html-italic">p</span> &lt; 0.05 in one-way ANOVA) relative to HDFa cells, and hashtags indicate statistically significant differences (at <span class="html-italic">p</span> &lt; 0.05 in two-way ANOVA and post hoc Tukey’s test) relative to non-treated MPS I or MPS II fibroblasts.</p>
Full article ">Figure 4
<p>Hsp70 and Hsp40 levels in brain and liver tissues of MPS I mice. Western blotting analyses (with GAPDH as a loading control) of the abundance of Hsp70 and Hsp40 in the brains and livers of MPS I (<span class="html-italic">Idua</span><sup>−/−</sup>) and control (<span class="html-italic">Idua</span><sup>+/+</sup>) mice. Representative blots are shown (<b>A</b>), and quantitative analyses (based on densitometry) from 6 animals in each group, with error bars representing SD, are demonstrated (<b>B</b>). Asterisks indicate statistically significant differences (at <span class="html-italic">p</span> &lt; 0.05 in two-way ANOVA and post hoc Tukey’s test) relative to control (<span class="html-italic">Idua</span><sup>+/+</sup>) mice.</p>
Full article ">Figure 5
<p>Changes in expression of genes coding for proteins involved in protein folding (<b>A</b>), response to endoplasmic reticulum stress (<b>B</b>), and response to unfolded protein (<b>C</b>) in MPS cells. Transcriptomic analyses indicating up- and down-regulation of genes represented in indicated Gene Ontology pathways (terms) in tested MPS fibroblasts relative to HDFa control cells. The number of genes of each class with significantly affected expression in MPS cells is shown in every panel.</p>
Full article ">Figure 6
<p>Number of transcripts included into indicated Gene Ontology terms with altered expression (<span class="html-italic">p</span> &lt; 0.1 in one-way ANOVA and post hoc Student’s <span class="html-italic">t</span>-test with Bonferroni correction) depending on the level of fold-change (FC) value in different types of MPS relative to HDFa control cells.</p>
Full article ">Figure 7
<p>Changes in protein processing in MPS cells. The KEGG pathway presenting the ‘protein processing in endoplasmic reticulum’ process, imaged from transcriptomic data derived from MPS cells. Individual proteins or groups of proteins were colored if a change in the expression of an indicated gene was observed in at least one type/subtype of MPS. Down-regulated and up-regulated genes are marked in blue and red, respectively. Non-marked values indicate results in which no statistically significant differences between MPS and HDFa were determined.</p>
Full article ">
Back to TopTop