Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Aug 14;25(16):8856.
doi: 10.3390/ijms25168856.

The Role of the Gut Microbiota in Sanfilippo Syndrome's Physiopathology: An Approach in Two Affected Siblings

Affiliations

The Role of the Gut Microbiota in Sanfilippo Syndrome's Physiopathology: An Approach in Two Affected Siblings

Raquel Barbero-Herranz et al. Int J Mol Sci. .

Abstract

Sanfilippo syndrome, or mucopolysaccharidosis type III (MPS III), is a rare lysosomal disease caused by congenital enzymatic deficiencies in heparan sulfate (HS) degradation, leading to organ dysfunction. The most severe hallmark of MPS III comprises neurological alterations, although gastrointestinal symptoms (GISs) have also been shown to be relevant in many patients. Here, we explored the contribution of the gut microbiota to MPS III GISs. We analyzed the composition and functionality of the gut microbiota in two MPS III siblings with the same mutation (c.544C > T, c.1080delC, in the SGSH gene) and the same diet, but with differences in their GISs, including recurrent diarrhea in one of them. Using 16S sequencing, we observed that the MPS III patients exhibited decreased alpha diversity and a lower abundance of Lachnospiraceae and Bifidobacteriaceae accompanied by a higher abundance of the Ruminococcaceae and Rikenellaceae families than the healthy control subjects. Comparing siblings, we found an increased abundance of Bacteroidaceae and a lower abundance of Ruminococcaceae and Akkermansiaceae in the GIS-free patient. This patient also had a higher relative abundance of Sus genes (SusA, SusB, SusE, and SusG) involved in glycosaminoglycan metabolism. We found higher HS levels in the stool of the two MPS III patients than in healthy volunteers, particularly in the patient with GISs. Functionally, whole fecal metabolites from the patient with GISs induced oxidative stress in vitro in healthy monocytes. Finally, the Bacteroides thetaiotaomicron strain isolated from MPS III stool samples exhibited HS degradation ability. Overall, our results reveal different microbiota compositions and functionalities in MPS III siblings, who exhibited differential gastrointestinal symptomatology. Our study may serve as a gateway to explore the impact of the gut microbiota and its potential to enhance the quality of life in Sanfilippo syndrome patients.

Keywords: Bacteroides thetaiotaomicron; SCFAs; Sanfilippo syndrome; Sus genes; gut microbiota.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Figures

Figure 1
Figure 1
Analysis of gut microbiota in healthy volunteers (HVs) and patients with Sanfilippo syndrome (MPS III). (A) The alpha diversity, Chao1, Shannon, and Faith PD indexes and the principal coordinates analysis (PCoA) plot of Bray−Curtis beta diversity are shown. Microbial composition was analyzed by 16S rRNA amplicon sequencing of feces from 4 HVs and the MPS III siblings included in the study. Relative abundance of families (B) and the 20 most abundant genera (C). f__, family; GIS, gastrointestinal symptoms; g__, genus.
Figure 2
Figure 2
Relative abundance of PUL (SusA, SusB, SusE, and SusG) genes assessed by qPCR from stool samples of MPS III siblings and healthy volunteers. GIS, gastrointestinal symptoms; HV, healthy volunteers; MPS III, Sanfilippo syndrome siblings. Each dot represents the average of 3 technical replicates and bars are expressed as mean ± SD. HV, n = 4 stool samples from 4 different healthy volunteers; GIS+, n = 2 two different day samples from the GIS+ MPS III patient.; GIS−, n = 2 two different day stool samples from the GIS+ MPS III patient.
Figure 3
Figure 3
Fecal levels of heparan sulfate (HS) (A), acetate (B), propionate (C), and butyrate (D) were determined by ELISA and gas chromatography coupled with mass spectrometry (GC-MS), respectively. Each dot represents the average of 3 technical replicates and bars are expressed as mean ± SD. HV, n = 4 stool samples from 4 different healthy volunteers; GIS+, n = 2 two different day samples from the GIS+ MPS III patient.; GIS−, n = 2 two different day stool samples from the GIS+ MPS III patient. GIS, gastrointestinal symptoms; HV, healthy volunteers; MPS III, Sanfilippo syndrome siblings.
Figure 4
Figure 4
Effects of MPS III fecal metabolites on healthy monocytes. Monocytes from 5 healthy volunteers’ peripheral blood were isolated and stimulated for 24 h with fecal metabolites from patients with MPS III. (A) Oxidative stress in gated monocytes established by Green CellROX staining was analyzed by means of flow cytometry. Levels of IL-1β (B), IL-6 (C), and TNF-α (D) in monocyte culture supernatant are shown. Each dot represents the average from experiments with 5 independent human healthy monocytes and bars are expressed as mean ± SD. HV, n = 4 stool samples from 4 different healthy volunteers; GIS+, n = 2 two different day stool samples from the GIS+ MPS III patient.; GIS−, n = 2 two different day samples from the GIS+ MPS III patient. Unst, unstimulated.
Figure 5
Figure 5
HS degradation activity of bacteria isolated from the stool of MPS III patients. Bacteroides spp. strains were isolated from stool samples of the two MPS III siblings included in this study. Bacteria were cultured in the presence of 75 µg/mL of HS at 37 °C under anaerobic conditions for 48 h. (A) Total GAG in the culture supernatant determined using the 1,9-dimethylmethylene blue procedure is shown. (B) Percentage of total GAG normalized to the PBS control. Bf, Bacteroides fragilis; Bt, Bacteroides thetaiotaomicron; n.d., not detected. KW, Kruskal–Wallis statistic; * p-value < 0.05 and *** p-value < 0.001 analyzed using the Kruskal–Wallis ANOVA test followed by Dunn’s multiple comparisons test. Data are expressed as mean ± SD from n = 4 independent experiments.

References

    1. Andrade F., Aldámiz-Echevarría L., Llarena M., Couce M.L. Sanfilippo Syndrome: Overall Review. Pediatr. Int. 2015;57:331–338. doi: 10.1111/ped.12636. - DOI - PubMed
    1. Heon-Roberts R., Nguyen A.L.A., Pshezhetsky A.V. Molecular Bases of Neurodegeneration and Cognitive Decline, the Major Burden of Sanfilippo Disease. J. Clin. Med. 2020;9:344. doi: 10.3390/jcm9020344. - DOI - PMC - PubMed
    1. Fecarotta S., Tarallo A., Damiano C., Minopoli N., Parenti G. Pathogenesis of Mucopolysaccharidoses, an Update. Int. J. Mol. Sci. 2020;21:2515. doi: 10.3390/ijms21072515. - DOI - PMC - PubMed
    1. Benetó N., Vilageliu L., Grinberg D., Canals I. Sanfilippo Syndrome: Molecular Basis, Disease Models and Therapeutic Approaches. Int. J. Mol. Sci. 2020;21:7819. doi: 10.3390/ijms21217819. - DOI - PMC - PubMed
    1. De Pasquale V., Pavone L.M. Heparan Sulfate Proteoglycans: The Sweet Side of Development Turns Sour in Mucopolysaccharidoses. Biochim. Biophys. Acta Mol. Basis Dis. 2019;1865:165539. doi: 10.1016/j.bbadis.2019.165539. - DOI - PubMed

Substances

LinkOut - more resources