26 research outputs found

    Inflammation-Associated Microbiota Composition Across Domestic Animals

    Get PDF
    Domestic animals represent important resources for understanding shared mechanisms underlying complex natural diseases that arise due to both genetic and environmental factors. Intestinal inflammation, particularly inflammatory bowel disease (IBD), is a significant health challenge in humans and domestic animals. While the etiology of IBD is multifactorial, imbalance of symbiotic gut microbiota has been hypothesized to play a central role in disease pathophysiology. Advances in genomic sequencing and analytical pipelines have enabled researchers to decipher the composition of the intestinal microbiota during health and in the context of naturally occurring diseases. This review compiles microbiome genomic data across domestic species and highlights a common occurrence of gut microbiome dysbiosis during idiopathic intestinal inflammation in multiple species, including dogs, cats, horses, cows, and pigs. Current microbiome data obtained from animals with intestinal inflammation are mostly limited to taxonomical analyses in association with broad clinical phenotype. In general, a pathogen or pathosymbiont were not detected. Rather, functional potential of the altered microbiota has been suggested to be one of the key etiologic factors. Among the domestic species studied, canine analyses are currently the most advanced with incorporation of functional profiling of microbiota. Canine IBD parallels features of the disease in humans, thus canines represent a strong natural model for human IBD. While deeper analyses of metagenomic data, coupled with host molecular analyses are needed, comparative studies across domestic species can reveal shared microbial alterations and regulatory mechanisms that will improve our understanding of intestinal inflammation in both animals and humans

    Microbiota Inhibit Epithelial Pathogen Adherence by Epigenetically Regulating C-Type Lectin Expression

    Get PDF
    Numerous bacterial pathogens infect the mammalian host by initially associating with epithelial cells that line the intestinal lumen. Recent work has revealed that commensal bacteria that reside in the intestine promote defense against pathogenic infection, however whether the microbiota direct host pathways that alter pathogen adherence is not well-understood. Here, by comparing germ-free mice, we identify that the microbiota decrease bacterial pathogen adherence and dampen epithelial expression of the cell surface glycoprotein C-type lectin 2e (Clec2e). Functional studies revealed that overexpression of this lectin promotes adherence of intestinal bacterial pathogens to mammalian cells. Interestingly, microbiota-sensitive downregulation of Clec2e corresponds with decreased histone acetylation of the Clec2e gene in intestinal epithelial cells. Histone deacetylation and transcriptional regulation of Clec2e depends on expression and recruitment of the histone deacetylase HDAC3. Thus, commensal bacteria epigenetically instruct epithelial cells to decrease expression of a C-type lectin that promotes pathogen adherence, revealing a novel mechanism for how the microbiota promote innate defense against infection

    IL-10–producing Tfh cells accumulate with age and link inflammation with age-related immune suppression

    Get PDF
    Aging results in profound immune dysfunction, resulting in the decline of vaccine responsiveness previously attributed to irreversible defects in the immune system. In addition to increased interleukin-6 (IL-6), we found aged mice exhibit increased systemic IL-10 that requires forkhead box P3–negative (FoxP3−), but not FoxP3+, CD4+T cells. Most IL-10–producing cells manifested a T follicular helper (Tfh) phenotype and required the Tfh cytokines IL-6 and IL-21 for their accrual, so we refer to them as Tfh10 cells. IL-21 was also required to maintain normal serum levels of IL-6 and IL-10. Notably, antigen-specific Tfh10 cells arose after immunization of aged mice, and neutralization of IL-10 receptor signaling significantly restored Tfh-dependent antibody responses, whereas depletion of FoxP3+ regulatory and follicular regulatory cells did not. Thus, these data demonstrate that immune suppression with age is reversible and implicate Tfh10 cells as an intriguing link between “inflammaging” and impaired immune responses with age

    Molecular and physiologic significance of the nuclear receptor corepressor-HDAC3 complex

    No full text
    Nuclear hormone receptors (NRs) are critical transcription factors in regulating development, differentiation, and metabolism. This work extends our understanding of the molecular mechanism by which NRs repress transcription and provides initial in vivo analyses into how the mechanism of repression translates into physiologic importance. Unliganded thyroid hormone receptor (TR) actively represses transcription via the nuclear receptor corepressor (N-CoR)/histone deacetylase 3 (HDAC3) complex. Here we report that SNF2H, the mammalian ISWI chromatin remodeling ATPase, is also critical for repression of a TR regulated gene. In fact, gene repression by TR involves the targeting of SNF2H to repressed genes by the HDAC activity of the N-CoR/HDAC3 complex. Secondly, to determine the role of the N-CoR/HDAC3 interaction in vivo, we generated C57BL/6 mice homozygous for a point mutation that dramatically decreases N-CoR\u27s ability to interact with HDAC3 and to function as a corepressor for TR. Interestingly, unlike the N-CoR knockout mice, the N-CoR DAD mutant mice are viable. The viability of the N-CoR DAD mutant mice will allow us to probe, for the first time, the specific biological functions of the N-CoR/HDAC3 interaction. Preliminary results suggest a role for N-CoR/HDAC3 in thyroid hormone and circadian biology, as well as the inflammatory response

    Molecular and physiologic significance of the nuclear receptor corepressor-HDAC3 complex

    No full text
    Nuclear hormone receptors (NRs) are critical transcription factors in regulating development, differentiation, and metabolism. This work extends our understanding of the molecular mechanism by which NRs repress transcription and provides initial in vivo analyses into how the mechanism of repression translates into physiologic importance. Unliganded thyroid hormone receptor (TR) actively represses transcription via the nuclear receptor corepressor (N-CoR)/histone deacetylase 3 (HDAC3) complex. Here we report that SNF2H, the mammalian ISWI chromatin remodeling ATPase, is also critical for repression of a TR regulated gene. In fact, gene repression by TR involves the targeting of SNF2H to repressed genes by the HDAC activity of the N-CoR/HDAC3 complex. Secondly, to determine the role of the N-CoR/HDAC3 interaction in vivo, we generated C57BL/6 mice homozygous for a point mutation that dramatically decreases N-CoR\u27s ability to interact with HDAC3 and to function as a corepressor for TR. Interestingly, unlike the N-CoR knockout mice, the N-CoR DAD mutant mice are viable. The viability of the N-CoR DAD mutant mice will allow us to probe, for the first time, the specific biological functions of the N-CoR/HDAC3 interaction. Preliminary results suggest a role for N-CoR/HDAC3 in thyroid hormone and circadian biology, as well as the inflammatory response

    Epigenomics and the Microbiota

    No full text

    The histone-binding code of nuclear receptor co-repressors matches the substrate specificity of histone deacetylase 3

    No full text
    Ligands for nuclear receptors facilitate the exchange of co-repressors for coactivators, leading to chromatin modifications that favour the activation of gene transcription. Here, we show that the repressed state of an endogenous retinoic acid-regulated gene is quickly re-established after ligand removal. As expected, repression is characterized by recruitment of N-CoR/SMRT–HDAC3 (histone deacetylase 3) co-repressor complexes, leading to local histone hypoacetylation. The achievement of the repressed state involves the ordered deacetylation of lysines in H4 tails. This order is determined by the inherent substrate specificity of HDAC3, and unexpectedly predicts the binding preference of N-CoR/SMRT for submaximally acetylated H4 tails. The match between the specificity of acetyl-histone deacetylation by HDAC3 and the histone-binding preference of N-CoR/SMRT allows the co-repressor complex to stabilize and propagate repression of nuclear hormone receptor gene targets

    A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism

    No full text
    Disruption of the circadian clock exacerbates metabolic diseases including obesity and diabetes. Here we show that histone deacetylase 3 (HDAC3) recruitment to the genome displays a circadian rhythm in mouse liver. Histone acetylation is inversely related to HDAC3 binding, and this rhythm is lost when HDAC3 is absent. Although amounts of HDAC3 are constant, its genomic recruitment in liver corresponds to the expression pattern of the circadian nuclear receptor Rev-erbα. Rev-erbα colocalizes with HDAC3 near genes regulating lipid metabolism, and deletion of HDAC3 or Rev-erbα in mouse liver causes hepatic steatosis. Thus, genomic recruitment of HDAC3 by Rev-erbα directs a circadian rhythm of histone acetylation and gene expression required for normal hepatic lipid homeostasis

    Microbiota-derived butyrate dampens linaclotide stimulation of the guanylate cyclase C pathway in patient-derived colonoids.

    Full text link
    BACKGROUND & AIMS: Disorders of gut-brain interaction (DGBI) are complex conditions that result in decreased quality of life and a significant cost burden. Linaclotide, a guanylin cyclase C (GCC) receptor agonist, is approved as a DGBI treatment. However, its efficacy has been limited and variable across DGBI patients. Microbiota and metabolomic alterations are noted in DGBI patients, provoking the hypothesis that the microbiota may impact the GCC response to current therapeutics. METHODS: Human-derived intestinal organoids were grown from pediatric DGBI, non-IBD colon biopsies (colonoids). Colonoids were treated with 250 nM linaclotide and assayed for cGMP to develop a model of GCC activity. Butyrate was administered to human colonoids overnight at a concentration of 1 mM. Colonoid lysates were analyzed for cGMP levels by ELISA. For the swelling assay, colonoids were photographed pre- and post-treatment and volume was measured using ImageJ. Principal coordinate analyses (PCoA) were performed on the Bray-Curtis dissimilarity and Jaccard distance to assess differences in the community composition of short-chain fatty acid (SCFA) producing microbial species in the intestinal microbiota from pediatric patients with IBS and healthy control samples. KEY RESULTS: Linaclotide treatment induced a significant increase in [cGMP] and swelling of patient-derived colonoids, demonstrating a human in vitro model of linaclotide-induced GCC activation. Shotgun sequencing analysis of pediatric IBS patients and healthy controls showed differences in the composition of commensal SCFA-producing bacteria. Butyrate exposure significantly dampened linaclotide-induced cGMP levels and swelling in patient-derived colonoids. CONCLUSIONS & INFERENCES: Patient-derived colonoids demonstrate that microbiota-derived butyrate can dampen human colonic responses to linaclotide. This study supports incorporation of microbiota and metabolomic assessment to improve precision medicine for DGBI patients.http://deepblue.lib.umich.edu/bitstream/2027.42/191988/2/Neurogastroenterology Motil - 2023 - Velez Lopez - Microbiota‐derived butyrate dampens linaclotide stimulation of the.pdfPublished versionDescription of Neurogastroenterology Motil - 2023 - Velez Lopez - Microbiota‐derived butyrate dampens linaclotide stimulation of the.pdf : Published versio
    corecore