Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 20513, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37993670

ABSTRACT

Disruption of intestinal epithelial functions is linked to Crohn disease (CD) pathogenesis. We identified a widespread reduction in the expression of long non-coding RNAs (lncRNAs) including LHFPL3-AS2 in the treatment-naïve CD ileum of the RISK pediatric cohort. We validated the reduction of LHFPL3-AS2 in adult CD and noted a further reduction in patients with more severe CD from the RISK cohort. LHFPL3-AS2 knockdown in Caco-2 cells robustly affected epithelial monolayer morphogenesis with markedly reduced confluency and spreading, showing atypical rounding, and clumping. mRNA-seq analysis of LHFPL3-AS2 knockdown cells highlighted the reduction of genes and pathways linked with apical polarity, actin bundles, morphogenesis, and the b-catenin-TCF4 complex. LHFPL3-AS2 knockdown significantly reduced the ability of cells to form an internal lumen within the 3-dimensional (3D) cyst model, with mislocalization of actin and adherent and tight junction proteins, affecting epithelial polarity. LHFPL3-AS2 knockdown also resulted in defective mitotic spindle formation and consequent reduction in epithelial proliferation. Altogether, we show that LHFPL3-AS2 reduction affects epithelial morphogenesis, polarity, mitotic spindle formation, and proliferation, which are key processes in maintaining epithelial homeostasis in CD. Reduced expression of LHFPL3-AS2 in CD patients and its further reduction with ileal ulceration outcome, emphasizes its significance in this context.


Subject(s)
Crohn Disease , RNA, Long Noncoding , Adult , Humans , Child , Caco-2 Cells , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Crohn Disease/genetics , Actins/genetics , Cell Proliferation/genetics , Ileum/metabolism , Cell Line, Tumor
2.
J Wound Care ; 32(7): 428-436, 2023 Jul 02.
Article in English | MEDLINE | ID: mdl-37405944

ABSTRACT

Nearly four decades after cultured epidermal autografts (CEA) were first used for the treatment of extensive burn wounds, the current gold standard treatment remains grafting healthy autologous skin from a donor site to the damaged areas, with current skin substitutes limited in their clinical use. We propose a novel treatment approach, using an electrospun polymer nanofibrous matrix (EPNM) applied on-site directly on the CEA-grafted areas. In addition, we propose a personalised treatment on hard-to-heal areas, in which we spray suspended autologous keratinocytes integrated with 3D EPNM applied on-site, directly onto the wound bed. This method enables the coverage of larger wound areas than possible with CEA. We present the case of a 26-year-old male patient with full-thickness burns covering 98% of his total body surface area (TBSA). We were able to show that this treatment approach resulted in good re-epithelialisation, seen as early as seven days post CEA grafting, with complete wound closure within three weeks, and to a lesser extent in areas treated with cell spraying. Moreover, in vitro experiments confirmed the feasibility of using keratinocytes embedded within the EPNM: cell and culture viability, identity, purity and potency were determined. These experiments show that the skin cells are viable and can proliferate within the EPNM. The results presented are of a promising novel strategy for the development of personalised wound treatment, integrating on-the-spot 'printed' EPNM with autologous skin cells, which will be applied at the bedside, over deep dermal wounds, to accelerate healing time and wound closure.


Subject(s)
Burns , Nanofibers , Adult , Humans , Male , Burns/surgery , Cells, Cultured , Keratinocytes , Skin , Skin Transplantation/methods , Transplantation, Autologous
3.
JCI Insight ; 8(14)2023 07 24.
Article in English | MEDLINE | ID: mdl-37261910

ABSTRACT

Ulcerative colitis (UC), Crohn's disease (CD), and celiac disease are prevalent intestinal inflammatory disorders with nonsatisfactory therapeutic interventions. Analyzing patient data-driven cohorts can highlight disease pathways and new targets for interventions. Long noncoding RNAs (lncRNAs) are attractive candidates, since they are readily targetable by RNA therapeutics, show relative cell-specific expression, and play key cellular functions. Uniformly analyzing gut mucosal transcriptomics from 696 subjects, we have highlighted lncRNA expression along the gastrointestinal (GI) tract, demonstrating that, in control samples, lncRNAs have a more location-specific expression in comparison with protein-coding genes. We defined dysregulation of lncRNAs in treatment-naive UC, CD, and celiac diseases using independent test and validation cohorts. Using the Predicting Response to Standardized Pediatric Colitis Therapy (PROTECT) inception UC cohort, we defined and prioritized lncRNA linked with UC severity and prospective outcomes, and we highlighted lncRNAs linked with gut microbes previously implicated in mucosal homeostasis. HNF1A-AS1 lncRNA was reduced in all 3 conditions and was further reduced in more severe UC form. Similarly, the reduction of HNF1A-AS1 ortholog in mice gut epithelia showed higher sensitivity to dextran sodium sulfate-induced colitis, which was coupled with alteration in the gut microbial community. These analyses highlight prioritized dysregulated lncRNAs that can guide future preclinical studies for testing them as potential targets.


Subject(s)
Celiac Disease , Colitis, Ulcerative , Crohn Disease , RNA, Long Noncoding , Animals , Mice , Colitis, Ulcerative/genetics , Crohn Disease/genetics , RNA, Long Noncoding/genetics , Celiac Disease/genetics , Transcriptome , Prospective Studies
4.
J Crohns Colitis ; 17(6): 960-971, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-36655602

ABSTRACT

BACKGROUND AND AIMS: Widespread dysregulation of long non-coding RNAs [lncRNAs] including a reduction in GATA6-AS1 was noted in inflammatory bowel disease [IBD]. We previously reported a prominent inhibition of epithelial mitochondrial functions in ulcerative colitis [UC]. However, the connection between reduction of GATA6-AS1 expression and attenuated epithelial mitochondrial functions was not defined. METHODS: Mucosal transcriptomics was used to conform GATA6-AS1 reduction in several treatment-naïve independent human cohorts [n=673]. RNA pull-down followed by mass spectrometry was used to determine the GATA6-AS1 interactome. Metabolomics and mitochondrial respiration following GATA6-AS1 silencing in Caco-2 cells were used to elaborate on GATA6-AS1 functions. RESULTS: GATA6-AS1 showed predominant expression in gut epithelia using single cell datasets. GATA6-AS1 levels were reduced in Crohn's disease [CD] ileum and UC rectum in independent cohorts. Reduced GATA6-AS1 lncRNA was further linked to a more severe UC form, and to a less favourable UC course. The GATA6-AS1 interactome showed robust enrichment for mitochondrial proteins, and included TGM2, an autoantigen in coeliac disease that is induced in UC, CD and coeliac disease, in contrast to GATA6-AS1 reduction in these cohorts. GATA6-AS1 silencing resulted in induction of TGM2, and this was coupled with a reduction in mitochondrial membrane potential and mitochondrial respiration, as well as in a reduction of metabolites linked to aerobic respiration relevant to mucosal inflammation. TGM2 knockdown in GATA6-AS1-deficient cells rescued mitochondrial respiration. CONCLUSIONS: GATA6-AS1 levels are reduced in UC, CD and coeliac disease, and in more severe UC forms. We highlight GATA6-AS1 as a target regulating epithelial mitochondrial functions, potentially through controlling TGM2 levels.


Subject(s)
Celiac Disease , Colitis, Ulcerative , Crohn Disease , Humans , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Caco-2 Cells , Intestinal Mucosa/metabolism , Crohn Disease/metabolism , Rectum , Inflammation/metabolism , Mitochondria/metabolism , GATA6 Transcription Factor/metabolism
5.
NPJ Biofilms Microbiomes ; 8(1): 2, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017536

ABSTRACT

The human gut microbiome develops during the first years of life, followed by a relatively stable adult microbiome. Day care attendance is a drastic change that exposes children to a large group of peers in a diverse environment for prolonged periods, at this critical time of microbial development, and therefore has the potential to affect microbial composition. We characterize the effect of day care on the gut microbial development throughout a single school year in 61 children from 4 different day care facilities, and in additional 24 age-matched home care children (n = 268 samples, median age of entering the study was 12 months). We show that day care attendance is a significant and impactful factor in shaping the microbial composition of the growing child, the specific daycare facility and class influence the gut microbiome, and each child becomes more similar to others in their day care. Furthermore, in comparison to home care children, day care children have a different gut microbial composition, with enrichment of taxa more frequently observed in older populations. Our results provide evidence that daycare may be an external factor that contributes to gut microbiome maturation and make-up in early childhood.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Adult , Aged , Child , Child, Preschool , Day Care, Medical , Humans , Infant
6.
Sci Rep ; 9(1): 16163, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31700112

ABSTRACT

Celiac disease is provoked by gluten exposure, but the complete pathogenic process in the duodenum and the loss of tolerance to gluten is not well understood. We aimed to define the core celiac transcriptomic signature and pathologic pathways in pre-treatment formalin-fixed paraffin-embedded (FFPE) duodenum biopsies used for clinical diagnosis. We use mRNAseq to define pre-treatment diagnostic duodenum gene expression in 54 pediatric celiac patients and non-celiac controls, and we validate our key findings in two independent cohorts of 67 adults and pediatric participants that used fresh frozen biopsies. We further define similar and divergent genes and pathways in 177 small bowel Crohn disease patients and controls. We observe a marked suppression of mature epithelial metabolic functions in celiac patients, overlapping substantially with the Crohn disease signature. A marked adaptive immune response was noted for the up-regulated signature including interferon response, alpha-beta, and gamma-delta T-cells that overlapped to some extent with the Crohn disease signature. However, we also identified a celiac disease specific signature linked to increased cell proliferation, nuclear division, and cell cycle activity that was localized primarily to the epithelia as noted by CCNB1 and Ki67 staining. Lastly, we demonstrate the utility of the transcriptomic date to correctly classify disease or healthy states in the discovery and validation cohorts. Our data supplement recently published datasets providing insights into celiac pathogenesis using clinical pathology FFPE samples, and can stimulate new approaches to address this highly prevalent condition.


Subject(s)
Celiac Disease , Duodenum , Intestinal Mucosa , Transcriptome , Adolescent , Biopsy , Celiac Disease/diagnosis , Celiac Disease/metabolism , Celiac Disease/pathology , Child , Child, Preschool , Crohn Disease/drug therapy , Crohn Disease/metabolism , Crohn Disease/pathology , Cyclin B1/biosynthesis , Duodenum/metabolism , Duodenum/pathology , Female , Gene Expression Regulation , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ki-67 Antigen/biosynthesis , Male
7.
Am J Gastroenterol ; 114(7): 1142-1151, 2019 07.
Article in English | MEDLINE | ID: mdl-30741738

ABSTRACT

OBJECTIVES: Crohn's disease (CD) is a chronic relapsing-remitting gut inflammatory disorder with a heterogeneous unpredictable course. Dysbiosis occurs in CD; however, whether microbial dynamics in quiescent CD are instrumental in increasing the risk of a subsequent flare remains undefined. METHODS: We analyzed the long-term dynamics of microbial composition in a prospective observational cohort of patients with quiescent CD (45 cases, 217 samples) over 2 years or until clinical flare occurred, aiming to identify whether changes in the microbiome precede and predict clinical relapse. Machine learning was used to prioritize microbial and clinical factors that discriminate between relapsers and nonrelapsers in the quiescent phase. RESULTS: Patients with CD in clinical, biomarker, and mucosal remission showed significantly reduced microbial richness and increased dysbiosis index compared with healthy controls. Of the 45 patients with quiescent CD, 12 (27%) flared during follow-up. Samples in quiescent patients preceding flare showed significantly reduced abundance of Christensenellaceae and S24.7, and increased abundance of Gemellaceae compared with those in remission throughout. A composite flare index was associated with a subsequent flare. Notably, higher individualized microbial instability in the quiescent phase was associated with a higher risk of a subsequent flare (hazard ratio 11.32, 95% confidence interval 3-42, P = 0.0035) using two preflare samples. Importantly, machine learning prioritized the flare index and the intrapersonal instability over clinical factors to best discriminate between relapsers and nonrelapsers. DISCUSSION: Individualized microbial variations in quiescent CD significantly increase the risk of future exacerbation and may provide a model to guide personalized preemptive therapy intensification.


Subject(s)
Crohn Disease/microbiology , Crohn Disease/pathology , Disease Progression , Dysbiosis/complications , Gastrointestinal Microbiome/physiology , Monitoring, Physiologic/methods , Adult , Case-Control Studies , Crohn Disease/therapy , Female , Follow-Up Studies , Humans , Intestinal Mucosa/microbiology , Linear Models , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Recurrence , Reference Values , Risk Assessment , Severity of Illness Index , Statistics, Nonparametric , Time Factors
8.
J Vis Exp ; (133)2018 03 19.
Article in English | MEDLINE | ID: mdl-29608151

ABSTRACT

The human intestinal microbiome plays a central role in protecting cells from injury, in processing energy and nutrients, and in promoting immunity. Deviations from what is considered a healthy microbiota composition (dysbiosis) may impair vital functions leading to pathologic conditions. Recent and ongoing research efforts have been directed toward the characterization of associations between microbial composition and human health and disease. Advances in high-throughput sequencing technologies enable characterization of the gut microbial composition. These methods include 16S rRNA-amplicon sequencing and shotgun sequencing. 16S rRNA-amplicon sequencing is used to profile taxonomical composition, while shotgun sequencing provides additional information about gene predictions and functional annotation. An advantage in using a targeted sequencing method of the 16S rRNA gene variable region is its substantially lower cost compared to shotgun sequencing. Sequence differences in the 16S rRNA gene are used as a microbial fingerprint to identify and quantify different taxa within an individual sample. Major international efforts have enlisted standards for 16S rRNA-amplicon sequencing. However, several studies report a common source of variation caused by batch effect. To minimize this effect, uniformed protocols for sample collection, processing, and sequencing must be implemented. This protocol proposes the integration of broadly used protocols starting from fecal sample collection to data analyses. This protocol includes a column-free, direct-PCR approach that enables simultaneous handling and DNA extraction of large numbers of fecal samples, along with PCR amplification of the V4 region. In addition, the protocol describes the analysis pipeline and provides a script using the latest version of QIIME (QIIME 2 version 2017.7.0 and DADA2). This step-by-step protocol is aimed to guide those interested in initiating the use of 16S rRNA-amplicon sequencing in a robust, reproductive, easy to use, detailed way.


Subject(s)
Feces/microbiology , High-Throughput Nucleotide Sequencing/methods , RNA, Ribosomal, 16S/genetics , Humans
9.
Inflamm Bowel Dis ; 24(2): 346-360, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29361088

ABSTRACT

Background: Long noncoding RNAs (lncRNA) are key regulators of gene transcription and many show tissue-specific expression. We previously defined a novel inflammatory and metabolic ileal gene signature in treatment-naive pediatric Crohn disease (CD). We now extend our analyses to include potential regulatory lncRNA. Methods: Using RNAseq, we systematically profiled lncRNAs and protein-coding gene expression in 177 ileal biopsies. Co-expression analysis was used to identify functions and tissue-specific expression. RNA in situ hybridization was used to validate expression. Real-time polymerase chain reaction was used to test lncRNA regulation by IL-1ß in Caco-2 enterocytes. Results: We characterize widespread dysregulation of 459 lncRNAs in the ileum of CD patients. Using only the lncRNA in discovery and independent validation cohorts showed patient classification as accurate as the protein-coding genes, linking lncRNA to CD pathogenesis. Co-expression and functional annotation enrichment analyses across several tissues and cell types 1showed that the upregulated LINC01272 is associated with a myeloid pro-inflammatory signature, whereas the downregulated HNF4A-AS1 exhibits association with an epithelial metabolic signature. We confirmed tissue-specific expression in biopsies using in situ hybridization, and validated regulation of prioritized lncRNA upon IL-1ß exposure in differentiated Caco-2 cells. Finally, we identified significant correlations between LINC01272 and HNF4A-AS1 expression and more severe mucosal injury. Conclusions: We systematically define differentially expressed lncRNA in the ileum of newly diagnosed pediatric CD. We show lncRNA utility to correctly classify disease or healthy states and demonstrate their regulation in response to an inflammatory signal. These lncRNAs, after mechanistic exploration, may serve as potential new tissue-specific targets for RNA-based interventions.


Subject(s)
Crohn Disease/genetics , Hepatocyte Nuclear Factor 4/genetics , RNA, Long Noncoding/genetics , Adolescent , Caco-2 Cells , Child , Down-Regulation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Ileum/metabolism , Ileum/pathology , Male , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Up-Regulation
10.
Sci Rep ; 7(1): 1088, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28439072

ABSTRACT

Hospitalized patients are at increased risk for acquiring healthcare-associated infections (HAIs) and inadequate nutrition. The human intestinal microbiota plays vital functions in nutrient supply and protection from pathogens, yet characterization of the microbiota of hospitalized patients is lacking. We used 16S rRNA amplicon sequencing to characterize the global pattern of microbial composition of fecal samples from 196 hospitalized patients with suspected infectious diarrhea in comparison to healthy, non-hospitalized subjects (n = 881), and to traditional culture results. We show that hospitalized patients have a significant rise in α-diversity (richness within sample) from birth to <4 years of age, which continues up to the second decade of life. Additionally, we noted a profoundly significant increase in taxa from Proteobacteria phylum in comparison to healthy subjects. Finally, although more than 60% of hospitalized samples had a greater than 10% abundance of Proteobacteria, there were only 19/196 (10%) positive cultures for Campylobacter, Salmonella, or Shigella entero-pathogens in traditional culturing methods. As hospitalized patients have increased risk for HAIs and inadequate nutrition, our data support the consideration of nutritional and/or microbial modification in this population.


Subject(s)
Bacteria/classification , Diarrhea/microbiology , Dysbiosis , Feces/microbiology , Microbiota , Adolescent , Adult , Aged , Bacteria/genetics , Child , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Female , Hospitalization , Humans , Male , Middle Aged , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Young Adult
11.
Article in English | MEDLINE | ID: mdl-26301853

ABSTRACT

Wastewater disposal may be a source of environmental contamination by Cryptosporidium and Giardia. This study was conducted to evaluate the prevalence of Cryptosporidium oocysts and Giardia cysts in raw and treated wastewater effluents. A prevalence of 100% was demonstrated for Giardia cysts in raw wastewater, at a concentration range of 10 to 12,225 cysts L(-1), whereas the concentration of Cryptosporidium oocysts in raw wastewater was 4 to 125 oocysts L(-1). The removal of Giardia cysts by secondary and tertiary treatment processes was greater than those observed for Cryptosporidium oocysts and turbidity. Cryptosporidium and Giardia were present in 68.5% and 76% of the tertiary effluent samples, respectively, at an average concentration of 0.93 cysts L(-1) and 9.94 oocysts L(-1). A higher detection limit of Cryptosporidium oocysts in wastewater was observed for nested PCR as compared to immune fluorescent assay (IFA). C. hominis was found to be the dominant genotype in wastewater effluents followed by C. parvum and C. andersoni or C. muris. Giardia was more prevalent than Cryptosporidium in the studied community and treatment processes were more efficient for the removal of Giardia than Cryptosporidium. Zoonotic genotypes of Cryptosporidium were also present in the human community. To assess the public health significance of Cryptosporidium oocysts present in tertiary effluent, viability (infectivity) needs to be assessed.


Subject(s)
Cryptosporidium/isolation & purification , Giardia/isolation & purification , Wastewater/parasitology , Water Purification/methods , Animals , Coloring Agents/isolation & purification , Cryptosporidium/cytology , Enterobacteriaceae/cytology , Enterobacteriaceae/isolation & purification , Feces/microbiology , Feces/parasitology , Giardia/cytology , Humans , Oocysts/cytology , Recycling/methods , Seasons
SELECTION OF CITATIONS
SEARCH DETAIL
...