Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 166991, 2024 03.
Article in English | MEDLINE | ID: mdl-38128843

ABSTRACT

Hirschsprung disease (HSCR) is a complex genetic disorder characterized by the absence of enteric nervous system (ENS) in the distal region of the intestine. Down Syndrome (DS) patients have a >50-fold higher risk of developing HSCR than the general population, suggesting that overexpression of human chromosome 21 (Hsa21) genes contribute to HSCR etiology. However, identification of responsible genes remains challenging. Here, we describe a genetic screening of potential candidate genes located on Hsa21, using the zebrafish. Candidate genes were located in the DS-HSCR susceptibility region, expressed in the human intestine, were known potential biomarkers for DS prenatal diagnosis, and were present in the zebrafish genome. With this approach, four genes were selected: RCAN1, ITSN1, ATP5PO and SUMO3. However, only overexpression of ATP5PO, coding for a component of the mitochondrial ATPase, led to significant reduction of ENS cells. Paradoxically, in vitro studies showed that overexpression of ATP5PO led to a reduction of ATP5PO protein levels. Impaired neuronal differentiation and reduced mitochondrial ATP production, were also detected in vitro, after overexpression of ATP5PO in a neuroblastoma cell line. Finally, epistasis was observed between ATP5PO and ret, the most important HSCR gene. Taken together, our results identify ATP5PO as the gene responsible for the increased risk of HSCR in DS patients in particular if RET variants are also present, and show that a balanced expression of ATP5PO is required for normal ENS development.


Subject(s)
Down Syndrome , Enteric Nervous System , Hirschsprung Disease , Animals , Humans , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Down Syndrome/genetics , Down Syndrome/metabolism , Zebrafish/genetics , Enteric Nervous System/metabolism , Biomarkers/metabolism
2.
Neurosci Biobehav Rev ; 136: 104634, 2022 05.
Article in English | MEDLINE | ID: mdl-35339484

ABSTRACT

Obesity is a complex disease with many co-morbidities, including impaired cognitive functions. Obese individuals often contain an aberrant microbiota. Via the microbiota-gut-brain axis, the altered microbiota composition can affect cognition or induce anxiety- or depressive-like behavior. Probiotics have been shown to alleviate both obesity- and neurobehavioral disorder-related symptoms. Here, we evaluated previously published results on the effectiveness of probiotic intervention in alleviating obesity- or high-fat diet (HFD)-related cognitive impairment, depression and anxiety. A systematic search was performed in PubMed, Embase, and Google Scholar until June 2021 to identify relevant articles. Seventeen studies were included: one human and sixteen animal studies. Overall, the findings support the beneficial health effect of probiotics on HFD-induced cognitive impairment and anxiety. However, the results suggest that multi-strain probiotic treatments should be used with caution, especially in the absence of HFD-induced impairment. Future studies should overcome the large variation in study design and high risk of bias found in the current evidence. Nevertheless, probiotic treatment, in particular using the Lactobacillus genus, seems promising.


Subject(s)
Cognitive Dysfunction , Probiotics , Animals , Anxiety/therapy , Cognitive Dysfunction/etiology , Depression/therapy , Diet, High-Fat/adverse effects , Humans , Obesity , Probiotics/pharmacology , Probiotics/therapeutic use
3.
Mol Med ; 26(1): 82, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32883198

ABSTRACT

BACKGROUND: Chorioamnionitis, inflammation of the chorion and amnion, which often results from intrauterine infection, is associated with premature birth and contributes to significant neonatal morbidity and mortality, including necrotizing enterocolitis (NEC). Recently, we have shown that chronic chorioamnionitis is associated with significant structural enteric nervous system (ENS) abnormalities that may predispose to later NEC development. Understanding time point specific effects of an intra-amniotic (IA) infection on the ENS is important for further understanding the pathophysiological processes and for finding a window for optimal therapeutic strategies for an individual patient. The aim of this study was therefore to gain insight in the longitudinal effects of intrauterine LPS exposure (ranging from 5 h to 15 days before premature delivery) on the intestinal mucosa, submucosa, and ENS in fetal lambs by use of a well-established translational ovine chorioamnionitis model. METHODS: We used an ovine chorioamnionitis model to assess outcomes of the fetal ileal mucosa, submucosa and ENS following IA exposure to one dose of 10 mg LPS for 5, 12 or 24 h or 2, 4, 8 or 15 days. RESULTS: Four days of IA LPS exposure causes a decreased PGP9.5- and S100ß-positive surface area in the myenteric plexus along with submucosal and mucosal intestinal inflammation that coincided with systemic inflammation. These changes were preceded by a glial cell reaction with early systemic and local gut inflammation. ENS changes and inflammation recovered 15 days after the IA LPS exposure. CONCLUSIONS: The pattern of mucosal and submucosal inflammation, and ENS alterations in the fetus changed over time following IA LPS exposure. Although ENS damage seemed to recover after prolonged IA LPS exposure, additional postnatal inflammatory exposure, which a premature is likely to encounter, may further harm the ENS and influence functional outcome. In this context, 4 to 8 days of IA LPS exposure may form a period of increased ENS vulnerability and a potential window for optimal therapeutic strategies.


Subject(s)
Autonomic Nervous System Diseases/diagnosis , Autonomic Nervous System Diseases/etiology , Chorioamnionitis/veterinary , Enteric Nervous System/pathology , Sheep Diseases/etiology , Animals , Biomarkers , Disease Models, Animal , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/etiology , Female , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Pregnancy , Premature Birth , Sheep
4.
J Pharm Policy Pract ; 13: 9, 2020.
Article in English | MEDLINE | ID: mdl-32377348

ABSTRACT

BACKGROUND: Substandard and falsified medicines, mainly prevalent in low and middle-income countries (LMICs), cause avoidable morbidity and mortality, and put at stake the performance of health systems. They may be prevented by an adequate implementation of pharmaceutical Quality Assurance (QA) guidelines, but unfortunately, most guidelines address upstream stakeholders and specialized staff in the supply chain. A multi-layered approach is needed, in order to empower the health workers at the point-of-care to proactively contribute to the fight against poor-quality medicines.Visual inspection is a simple technique, suitable for field screening. The findings of a survey conducted in the Democratic Republic of the Congo (DRC) suggested that it might be a fairly good (yet partial) predictor of poor-quality, when compared to full laboratory tests. METHODS AND RESULTS: Starting from the 68-questions checklist originally used in the survey in the DRC, we developed a simplified checklist, specifically designed to guide health workers at the point of care to rapidly identify suspect poor-quality medicines. We selected those medicines' attributes the assessment of which does not require technical expertise, or access to regulatory information. Attributes were categorized according to a 3-level risk scale, to guide decision-making on suspect poor-quality medicines, based on an informed risk assessment.The simplified checklist contains 26 binary questions (YES/NO), grouped into four themes: packaging, identification, traceability, and physical appearance. Each non-conformity corresponds to a level of risk for patients. The user is guided towards three possible actions: A) reasonably safe for dispensing; B) dispense with explanation; C) quarantine and make a risk-benefit evaluation before dispensing. CONCLUSION: The simplified checklist should now be implemented in real-life setting in LMICs. If proven useful in guiding health workers at the point-of-care to take rapid, transparent, patient-centred actions when facing a suspect poor-quality medicine, it could be further extended to address specific formulations. Digitalization for linkage with pharmacovigilance programs could also be considered.

5.
Am J Gastroenterol ; 114(12): 1909-1918, 2019 12.
Article in English | MEDLINE | ID: mdl-31764091

ABSTRACT

INTRODUCTION: We set out to evaluate the performance of a multitarget stool DNA (MT-sDNA) in an average-risk colonoscopy-controlled colorectal cancer (CRC) screening population. MT-sDNA stool test results were evaluated against fecal immunochemical test (FIT) results for the detection of different lesions, including molecularly defined high-risk adenomas and several other tumor characteristics. METHODS: Whole stool samples (n = 1,047) were prospectively collected and subjected to an MT-sDNA test, which tests for KRAS mutations, NDRG4 and BMP3 promoter methylation, and hemoglobin. Results for detecting CRC (n = 7), advanced precancerous lesions (advanced adenoma [AA] and advanced serrated polyps; n = 119), and non-AAs (n = 191) were compared with those of FIT alone (thresholds of 50, 75, and 100 hemoglobin/mL). AAs with high risk of progression were defined by the presence of specific DNA copy number events as measured by low-pass whole genome sequencing. RESULTS: The MT-sDNA test was more sensitive than FIT alone in detecting advanced precancerous lesions (46% (55/119) vs 27% (32/119), respectively, P < 0.001). Specificities among individuals with nonadvanced or negative findings (controls) were 89% (791/888) and 93% (828/888) for MT-sDNA and FIT testing, respectively. A positive MT-sDNA test was associated with multiple lesions (P = 0.005), larger lesions (P = 0.03), and lesions with tubulovillous architecture (P = 0.04). The sensitivity of the MT-sDNA test or FIT in detecting individuals with high-risk AAs (n = 19) from individuals with low-risk AAs (n = 52) was not significantly different. DISCUSSION: In an average-risk screening population, the MT-sDNA test has an increased sensitivity for detecting advanced precancerous lesions compared with FIT alone. AAs with a high risk of progression were not detected with significantly higher sensitivity by MT-sDNA or FIT.


Subject(s)
Adenoma/diagnosis , Colonic Polyps/diagnosis , Colorectal Neoplasms/diagnosis , DNA/analysis , Feces/chemistry , Hemoglobins/analysis , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Aged , Bone Morphogenetic Protein 3/genetics , Colonic Polyps/genetics , Colonic Polyps/metabolism , Colonic Polyps/pathology , Colonoscopy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Early Detection of Cancer , Female , Hemoglobins/metabolism , Humans , Immunochemistry , Male , Middle Aged , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics
6.
Eur J Cancer ; 94: 104-114, 2018 05.
Article in English | MEDLINE | ID: mdl-29550565

ABSTRACT

BACKGROUND: Oesophageal (OeC) and gastric (GC) cancer patients are treated with similar multimodal therapy and have poor survival. There remains an urgent clinical need to identify biomarkers to individualise patient management and improve outcomes. Therapy with immune checkpoint inhibitors has shown promising results in other cancers. Proposed biomarkers to predict potential response to immune checkpoint inhibitors include DNA mismatch repair (MMR) and/or Epstein-Barr virus (EBV) status. The aim of this study was to establish and compare EBV status and MMR status in large multi-centre series of OeC and GC. METHODS: EBV was assessed by EBV-encoded RNA (EBER) in situ hybridisation and MMR protein expression by immunohistochemistry (IHC) in 988 OeC and 1213 GC from multiple centres. In a subset of OeC, microsatellite instability (MSI) was tested in parallel with MMR IHC. RESULTS: Frequency of MMR deficiency (MMRdef) and MSI was low in OeC (0.8% and 0.6%, respectively) compared with GC (10.3%). None of the OeCs were EBER positive in contrast to 4.8% EBER positive GC. EBV positive GC patients were younger (p = 0.01), more often male (p = 0.001) and had a better overall survival (p = 0.012). MMRdef GC patients were older (p = 0.001) and showed more often intestinal-type histology (p = 0.022). CONCLUSIONS: This is the largest study to date indicating that EBV and MMRdef do not play a role in OeC carcinogenesis in contrast to GC. The potential clinical usefulness of determining MMRdef/EBV status to screen patients for eligibility for immune-targeting therapy differs between OeC and GC patients.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Neoplasms/virology , Stomach Neoplasms/genetics , Stomach Neoplasms/virology , Adult , Aged , Biomarkers, Tumor/analysis , DNA Mismatch Repair/genetics , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/epidemiology , Female , Humans , Male , Microsatellite Instability , Middle Aged
7.
Article in English | MEDLINE | ID: mdl-28524415

ABSTRACT

BACKGROUND: Promoter methylation of N-myc Downstream-Regulated Gene 4 (NDRG4) in fecal DNA is an established early detection marker for colorectal cancer (CRC). Despite its connection to CRC, NDRG4 is predominantly studied in brain and heart, with little to no knowledge about its expression or role in other organs. In this study, we aimed to determine the whole-body expression of NDRG4, with a focus on the intestinal tract. METHODS: We investigated NDRG4 expression throughout the body by immunohistochemistry, Western Blotting and in situ mRNA hybridization using tissues from NDRG4 wild-type, heterozygous and knockout mice and humans. In addition, we explored cell-specific expression of NDRG4 in murine whole-mount gut preparations using immunofluorescence and confocal microscopy. KEY RESULTS: NDRG4 is specifically expressed within nervous system structures throughout the body. In the intestinal tract of both mouse and man, NDRG4 immunoreactivity was restricted to the enteric nervous system (ENS), where it labeled cell bodies of the myenteric and submucosal plexuses and interconnecting nerve fibers. More precisely, NDRG4 expression was limited to neurons, as NDRG4 always co-localized with HuC/D (pan-neuronal marker) but never with GFAP (an enteric glial cell marker). Furthermore, NDRG4 was expressed in various neuropeptide Y positive neurons, but was only found in a minority (~10%) of neurons expressing neuronal nitric oxide synthase. CONCLUSIONS AND INFERENCES: NDRG4 is exclusively expressed by central, peripheral and enteric neurons/nerves, suggesting a neuronal-specific role of this protein. Our findings raise the question whether NDRG4, via the ENS, an understudied component of the tumor microenvironment, supports CRC development and/or progression.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/metabolism , Enteric Nervous System/metabolism , Muscle Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/analysis , Nerve Tissue Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...