Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Front Pharmacol ; 15: 1409683, 2024.
Article in English | MEDLINE | ID: mdl-39050748

ABSTRACT

The gastrointestinal tract is lined by an epithelial monolayer responsible for selective permeability and absorption, as well as protection against harmful luminal contents. Recognition of foreign or aberrant DNA within these epithelial cells is, in part, regulated by pattern recognition receptors such as cyclic GMP-AMP synthase (cGAS). cGAS binds double-stranded DNA from exogenous and endogenous sources, resulting in the activation of stimulator of interferon genes (STING) and a type 1 interferon response. cGAS is also implicated in non-canonical pathways involving the suppression of DNA repair and the upregulation of autophagy via interactions with PARP1 and Beclin-1, respectively. The importance of cGAS activation in the development and progression of inflammatory bowel disease and gastrointestinal cancers has been and continues to be explored. This review delves into the intricacies of the complex role of cGAS in intestinal epithelial inflammation and gastrointestinal malignancies, as well as recent therapeutic advances targeting cGAS pathways.

2.
Front Immunol ; 14: 1231700, 2023.
Article in English | MEDLINE | ID: mdl-37744380

ABSTRACT

Introduction: We have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downregulation of PGC1α expression and activity during IBD is not yet clear. Methods: Mice (male; C57Bl/6, Villincre/+;Pgc1afl/fl mice, and Pgc1afl/fl) were subjected to experimental colitis and treated with nicotinamide riboside. Western blot, high-resolution respirometry, nicotinamide adenine dinucleotide (NAD+) quantification, and immunoprecipitation were used to in this study. Results: We demonstrate a significant depletion in the NAD+ levels within the intestinal epithelium of mice undergoing experimental colitis, as well as humans with ulcerative colitis. While we found no decrease in the levels of NAD+-synthesizing enzymes within the intestinal epithelium of mice undergoing experimental colitis, we did find an increase in the mRNA level, as well as the enzymatic activity, of the NAD+-consuming enzyme poly(ADP-ribose) polymerase-1 (PARP1). Treatment of mice undergoing experimental colitis with an NAD+ precursor reduced the severity of colitis, restored mitochondrial function, and increased active PGC1α levels; however, NAD+ repletion did not benefit transgenic mice that lack PGC1α within the intestinal epithelium, suggesting that the therapeutic effects require an intact PGC1α axis. Discussion: Our results emphasize the importance of PGC1α expression to both mitochondrial health and homeostasis within the intestinal epithelium and suggest a novel therapeutic approach for disease management. These findings also provide a mechanistic basis for clinical trials of nicotinamide riboside in IBD patients.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Male , Animals , Mice , NAD , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Mice, Transgenic , Mitochondria , Inflammation
3.
J Vis Exp ; (196)2023 06 02.
Article in English | MEDLINE | ID: mdl-37335108

ABSTRACT

The intestinal epithelium plays an essential role in human health, providing a barrier between the host and the external environment. This highly dynamic cell layer provides the first line of defense between microbial and immune populations and helps to modulate the intestinal immune response. Disruption of the epithelial barrier is a hallmark of inflammatory bowel disease (IBD) and is of interest for therapeutic targeting. The 3-dimensional colonoid culture system is an extremely useful in vitro model for studying intestinal stem cell dynamics and epithelial cell physiology in IBD pathogenesis. Ideally, establishing colonoids from the inflamed epithelial tissue of animals would be most beneficial in assessing the genetic and molecular influences on disease. However, we have shown that in vivo epithelial changes are not necessarily retained in colonoids established from mice with acute inflammation. To address this limitation, we have developed a protocol to treat colonoids with a cocktail of inflammatory mediators that are typically elevated during IBD. While this system can be applied ubiquitously to various culture conditions, this protocol emphasizes treatment on both differentiated colonoids and 2-dimensional monolayers derived from established colonoids. In a traditional culture setting, colonoids are enriched with intestinal stem cells, providing an ideal environment to study the stem cell niche. However, this system does not allow for an analysis of the features of intestinal physiology, such as barrier function. Further, traditional colonoids do not offer the opportunity to study the cellular response of terminally differentiated epithelial cells to proinflammatory stimuli. The methods presented here provide an alternative experimental framework to address these limitations. The 2-dimensional monolayer culture system also offers an opportunity for therapeutic drug screening ex vivo. This polarized layer of cells can be treated with inflammatory mediators on the basal side of the cell and concomitantly with putative therapeutics apically to determine their utility in IBD treatment.


Subject(s)
Colon , Inflammatory Bowel Diseases , Mice , Animals , Humans , Intestines/pathology , Intestinal Mucosa/metabolism , Inflammatory Bowel Diseases/pathology , Epithelial Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism
4.
FASEB J ; 36(5): e22282, 2022 05.
Article in English | MEDLINE | ID: mdl-35344224

ABSTRACT

Inflammatory bowel disease (IBD) represents a set of idiopathic and chronic inflammatory diseases of the gastrointestinal tract. Central to the pathogenesis of IBD is a dysregulation of normal intestinal epithelial homeostasis. cGAS is a DNA-sensing receptor demonstrated to promote autophagy, a mechanism that removes dysfunctional cellular components. Beclin-1 is a crucial protein involved in the initiation of autophagy. We hypothesized that cGAS plays a key role in intestinal homeostasis by upregulating Beclin-1-mediated autophagy. We evaluated intestinal cGAS levels in humans with IBD and in murine colonic tissue after performing a 2% dextran sulfate sodium (DSS) colitis model. Autophagy and cell death mechanisms were studied in cGAS KO and WT mice via qPCR, WB analysis, H&E, IF, and TUNEL staining. Autophagy was measured in stimulated intestinal epithelial cells (IECs) via WB analysis. Our data demonstrates cGAS to be upregulated during human and murine colitis. Furthermore, cGAS deficiency leads to worsened colitis and decreased levels of autophagy proteins including Beclin-1 and LC3-II. Co-IP demonstrates a direct binding between cGAS and Beclin-1 in IECs. Transfection of cGAS in stimulated HCT-116 cells leads to increased autophagy. IECs isolated from cGAS KO have diminished autophagic flux. cGAS KO mice subjected to DSS have increased cell death and cleaved caspase-3. Lastly, treatment of cGAS KO mice with rapamycin decreased the severity of colitis. Our data suggest that cGAS maintains intestinal epithelial homeostasis during human IBD and murine colitis by upregulating Beclin-1-mediated autophagy and preventing IEC death. Rescue of autophagy can attenuate the severity of colitis associated with cGAS deficiency.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Autophagy/physiology , Beclin-1/genetics , Colitis/metabolism , Dextran Sulfate/toxicity , Homeostasis , Inflammation/metabolism , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Nucleotides, Cyclic , Nucleotidyltransferases/genetics
5.
Inflamm Bowel Dis ; 28(7): 988-1003, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35259271

ABSTRACT

BACKGROUND: Perturbagen analysis of Crohn's disease (CD) ileal gene expression data identified small molecules including eicosatetraynoic acid (ETYA), which may exert an antifibrotic effect. We developed a patient-specific human intestinal organoid (HIO) model system to test small molecule regulation of mitochondrial and wound-healing functions implicated in stricturing behavior. METHODS: HIOs were made from CD induced pluripotent stem cells with and without a loss-of-function haplotype in the DUOX2 gene implicated in ileal homeostasis and characterized under basal conditions and following exposure to butyrate and ETYA using RNA sequencing, flow cytometry, and immunofluorescent and polarized light microscopy. Mitochondrial activity was measured using high-resolution respirometry and tissue stiffness using atomic force microscopy. RESULTS: HIOs expressed core mitochondrial and extracellular matrix (ECM) genes and enriched biologic functions implicated in CD ileal strictures; ECM gene expression was suppressed by both butyrate and ETYA, with butyrate also suppressing genes regulating epithelial proliferation. Consistent with this, butyrate, but not ETYA, exerted a profound effect on HIO epithelial mitochondrial function, reactive oxygen species production, and cellular abundance. Butyrate and ETYA suppressed HIO expression of alpha smooth muscle actin expressed by myofibroblasts, type I collagen, and collagen protein abundance. HIOs exhibited tissue stiffness comparable to normal human ileum; this was reduced by chronic ETYA exposure in HIOs carrying the DUOX2 loss-of-function haplotype. CONCLUSIONS: ETYA regulates ECM genes implicated in strictures and suppresses collagen content and tissue stiffness in an HIO model. HIOs provide a platform to test personalized therapeutics, including small molecules prioritized by perturbagen analysis.


A subset of pediatric Crohn's disease patients develop intestinal strictures requiring surgery. The microbial metabolite butyrate and eicosatetraynoic acid regulate pathways implicated in stricture formation in a human intestinal organoid model system, which may be used to test new therapies.


Subject(s)
Crohn Disease , Butyrates/metabolism , Butyrates/pharmacology , Collagen/metabolism , Constriction, Pathologic/metabolism , Crohn Disease/genetics , Dual Oxidases/metabolism , Extracellular Matrix/metabolism , Humans , Intestinal Mucosa/metabolism , Mitochondria/metabolism , Organoids/metabolism
6.
Adv Health Sci Educ Theory Pract ; 27(1): 167-187, 2022 03.
Article in English | MEDLINE | ID: mdl-34709484

ABSTRACT

PURPOSE: To perform a scoping review to determine what is known about emotional intelligence (EI) in undergraduate medical education (UME). Two main questions were asked: A. What medical student characteristics are associated with EI? Are there correlations with demographic or other factors? B. What research studies have been done on EI in UME? For example, is there evidence EI changes over time as a result of personal experiences? Should EI be used as an admission criterion? Can EI improve as a result of experiences or deliberate interventions? METHOD: The authors searched four databases (PubMed, PsycInfo, Education Resources Information Center, and Web of Science) for all papers published up to and including December 2020. Two reviewers independently screened articles to determine if they met inclusion criteria. All authors extracted and analyzed data. RESULTS: A set of 1520 papers on the topic of emotional intelligence was identified, with 119 papers meeting inclusion criteria. Most studies were done at international locations with only 17 done at US medical schools. Seventy-five were cohort or cross-sectional studies. Study populations were mixed among the studies, with year of medical study, inclusion of other healthcare students, and participation rates among the inter-study differences noted. CONCLUSIONS: Numerous gaps in the literature on EI exist with several points being clear: (1) there is disagreement on the definition of EI, (2) it is undetermined whether EI is a trait or an ability, and (3) there is marked variability among the instruments used to measure EI. It is also becoming apparent that using EI determination may be helpful as a component of the admission process, higher EI is likely related to improved clinical reasoning, and higher EI contributes to more effective stress management.


Subject(s)
Education, Medical, Undergraduate , Students, Medical , Cross-Sectional Studies , Emotional Intelligence , Humans , Students, Medical/psychology , Surveys and Questionnaires
7.
Antioxid Redox Signal ; 33(1): 1-19, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32103677

ABSTRACT

Aims: Mitochondrial stress and dysfunction within the intestinal epithelium are known to contribute to the pathogenesis of inflammatory bowel disease (IBD). However, the importance of mitophagy during intestinal inflammation remains poorly understood. The primary aim of this study was to investigate how the mitophagy protein BCL2/adenovirus E1B 19 kDa protein-interacting protein 3-like (BNIP3L/NIX) mitigates mitochondrial damage during intestinal inflammation in the hopes that these data will allow us to target mitochondrial health in the intestinal epithelium as an adjunct to immune-based treatment strategies. Results: In the intestinal epithelium of patients with ulcerative colitis, we found that NIX was upregulated and targeted to the mitochondria. We obtained similar findings in wild-type mice undergoing experimental colitis. An increase in NIX expression was found to depend on stabilization of hypoxia-inducible factor-1 alpha (HIF1α), which binds to the Nix promoter region. Using the reactive oxygen species (ROS) scavenger MitoTEMPO, we were able to attenuate disease and inhibit both HIF1α stabilization and subsequent NIX expression, suggesting that mitochondrially derived ROS are crucial to initiating the mitophagic response during intestinal inflammation. We subjected a global Nix-/- mouse to dextran sodium sulfate colitis and found that these mice developed worse disease. In addition, Nix-/- mice were found to exhibit increased mitochondrial mass, likely due to the inability to clear damaged or dysfunctional mitochondria. Innovation: These results demonstrate the importance of mitophagy within the intestinal epithelium during IBD pathogenesis. Conclusion: NIX-mediated mitophagy is required to maintain intestinal homeostasis during inflammation, highlighting the impact of mitochondrial damage on IBD progression.


Subject(s)
Gastroenteritis/etiology , Membrane Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitophagy/genetics , Animals , Antioxidants/pharmacology , Binding Sites , Biomarkers , Cell Line, Tumor , Colitis/etiology , Colitis/metabolism , Colitis/pathology , Cyclic N-Oxides/metabolism , Disease Models, Animal , Disease Susceptibility , Gastroenteritis/metabolism , Gastroenteritis/pathology , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Models, Biological , Promoter Regions, Genetic , Protein Binding , Reactive Oxygen Species/metabolism , Response Elements
8.
PLoS One ; 14(12): e0224838, 2019.
Article in English | MEDLINE | ID: mdl-31790417

ABSTRACT

The gut plays a vital role in critical illness, and alterations in the gut structure and function have been reported in endotoxemia and sepsis models. Previously, we have demonstrated a novel link between the diet-induced alteration of the gut microbiome with cellulose and improved outcomes in sepsis. As compared to mice receiving basal fiber (BF) diet, mice that were fed a non-fermentable high fiber (HF) diet demonstrated significant improvement in survival and decreased organ injury in both cecal-ligation and puncture (CLP) and endotoxin sepsis models. To understand if the benefit conferred by HF diet extends to the gut structure and function, we hypothesized that HF diet would be associated with a reduction in sepsis-induced gut epithelial loss and permeability in mice. We demonstrate that the use of dietary cellulose decreased LPS-mediated intestinal hyperpermeability and protected the gut from apoptosis. Furthermore, we noted a significant increase in epithelial cell proliferation, as evidenced by an increase in the percentage of bromodeoxyuridine-positive cells in HF fed mice as compared to BF fed mice. Thus, the use of HF diet is a simple and effective tool that confers benefit in a murine model of sepsis, and understanding the intricate relationship between the epithelial barrier, gut microbiota, and diet will open-up additional therapeutic avenues for the treatment of gut dysfunction in critical illness.


Subject(s)
Apoptosis/drug effects , Cellulose/pharmacology , Dietary Supplements , Endotoxemia/metabolism , Endotoxemia/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Endotoxemia/microbiology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Permeability/drug effects , Tight Junction Proteins/metabolism
10.
Nat Commun ; 10(1): 38, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604764

ABSTRACT

Molecular mechanisms driving disease course and response to therapy in ulcerative colitis (UC) are not well understood. Here, we use RNAseq to define pre-treatment rectal gene expression, and fecal microbiota profiles, in 206 pediatric UC patients receiving standardised therapy. We validate our key findings in adult and paediatric UC cohorts of 408 participants. We observe a marked suppression of mitochondrial genes and function across cohorts in active UC, and that increasing disease severity is notable for enrichment of adenoma/adenocarcinoma and innate immune genes. A subset of severity genes improves prediction of corticosteroid-induced remission in the discovery cohort; this gene signature is also associated with response to anti-TNFα and anti-α4ß7 integrin in adults. The severity and therapeutic response gene signatures were in turn associated with shifts in microbes previously implicated in mucosal homeostasis. Our data provide insights into UC pathogenesis, and may prioritise future therapies for nonresponders to current approaches.


Subject(s)
Colitis, Ulcerative/genetics , Genes, Mitochondrial/genetics , Intestinal Mucosa/metabolism , Mitochondrial Diseases/genetics , Transcriptome/genetics , Adolescent , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Child , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Feces/microbiology , Female , Gene Expression Profiling , Glucocorticoids/therapeutic use , Humans , Integrins/antagonists & inhibitors , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mesalamine/therapeutic use , Microbiota , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/microbiology , Mitochondrial Diseases/pathology , Precision Medicine/methods , Prospective Studies , Rectum/metabolism , Rectum/microbiology , Rectum/pathology , Remission Induction/methods , Sequence Analysis, RNA , Severity of Illness Index , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
FASEB J ; 33(1): 1330-1346, 2019 01.
Article in English | MEDLINE | ID: mdl-30113881

ABSTRACT

The incidence and prevalence of inflammatory bowel disease (IBD) are increasing worldwide. IBD is known to be multifactorial, but inflammatory signaling within the intestinal epithelium and a subsequent failure of the intestinal epithelial barrier have been shown to play essential roles in disease pathogenesis. CaMKIV is a multifunctional protein kinase associated with inflammation and cell cycle regulation. CaMKIV has been extensively studied in autoimmune diseases, but a role in idiopathic intestinal inflammation has not been described. In this study, active CaMKIV was highly expressed within the intestinal epithelium of humans with ulcerative colitis and wild-type (WT) mice with experimental induced colitis. Clinical disease severity directly correlates with CaMKIV activation, as does expression of proinflammatory cytokines and histologic features of colitis. In WT mice, CaMKIV activation is associated with increases in expression of 2 cell cycle proarrest signals: p53 and p21. Cell cycle arrest inhibits proliferation of the intestinal epithelium and ultimately results in compromised intestinal epithelial barrier integrity, further perpetuating intestinal inflammation during experimental colitis. Using a CaMKIV null mutant mouse, we demonstrate that a loss of CaMKIV protects against murine DSS colitis. Small molecules targeting CaMKIV activation may provide therapeutic benefit for patients with IBD.-Cunningham, K. E., Novak, E. A., Vincent, G., Siow, V. S., Griffith, B. D., Ranganathan, S., Rosengart, M. R., Piganelli, J. D., Mollen, K. P. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) activation contributes to the pathogenesis of experimental colitis via inhibition of intestinal epithelial cell proliferation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cell Proliferation , Colitis/enzymology , Colitis/pathology , Intestinal Mucosa/pathology , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Colitis/chemically induced , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Dextran Sulfate/toxicity , Enzyme Activation , Humans , Intestinal Mucosa/enzymology , Mice , Mice, Knockout , Signal Transduction
12.
Antioxidants (Basel) ; 6(4)2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29104232

ABSTRACT

Oxidative stress and persistent inflammation are exaggerated through chronic over-nutrition and a sedentary lifestyle, resulting in insulin resistance. In type 2 diabetes (T2D), impaired insulin signaling leads to hyperglycemia and long-term complications, including metabolic liver dysfunction, resulting in non-alcoholic fatty liver disease (NAFLD). The manganese metalloporphyrin superoxide dismustase (SOD) mimetic, manganese (III) meso-tetrakis (N-ethylpyridinium-2-yl) porphyrin (MnP), is an oxidoreductase known to scavenge reactive oxygen species (ROS) and decrease pro-inflammatory cytokine production, by inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. We hypothesized that targeting oxidative stress-induced inflammation with MnP would assuage liver complications and enhance insulin sensitivity and glucose tolerance in a high-fat diet (HFD)-induced mouse model of T2D. During 12 weeks of feeding, we saw significant improvements in weight, hepatic steatosis, and biomarkers of liver dysfunction with redox modulation by MnP treatment in HFD-fed mice. Additionally, MnP treatment improved insulin sensitivity and glucose tolerance, while reducing serum insulin and leptin levels. We attribute these effects to redox modulation and inhibition of hepatic NF-κB activation, resulting in diminished ROS and pro-inflammatory cytokine production. This study highlights the importance of controlling oxidative stress and secondary inflammation in obesity-mediated insulin resistance and T2D. Our data confirm the role of NF-κB-mediated inflammation in the development of T2D, and demonstrate the efficacy of MnP in preventing the progression to disease by specifically improving liver pathology and hepatic insulin resistance in obesity.

13.
PLoS One ; 12(4): e0175549, 2017.
Article in English | MEDLINE | ID: mdl-28426686

ABSTRACT

The immune system is necessary for protecting against various pathogens. However, under certain circumstances, self-reactive immune cells can drive autoimmunity, like that exhibited in type 1 diabetes (T1D). CD4+ T cells are major contributors to the immunopathology in T1D, and in order to drive optimal T cell activation, third signal reactive oxygen species (ROS) must be present. However, the role ROS play in mediating this process remains to be further understood. Recently, cellular metabolic programs have been shown to dictate the function and fate of immune cells, including CD4+ T cells. During activation, CD4+ T cells must transition metabolically from oxidative phosphorylation to aerobic glycolysis to support proliferation and effector function. As ROS are capable of modulating cellular metabolism in other models, we sought to understand if blocking ROS also regulates CD4+ T cell activation and effector function by modulating T cell metabolism. To do so, we utilized an ROS scavenging and potent antioxidant manganese metalloporphyrin (MnP). Our results demonstrate that redox modulation during activation regulates the mTOR/AMPK axis by maintaining AMPK activation, resulting in diminished mTOR activation and reduced transition to aerobic glycolysis in diabetogenic splenocytes. These results correlated with decreased Myc and Glut1 upregulation, reduced glucose uptake, and diminished lactate production. In an adoptive transfer model of T1D, animals treated with MnP demonstrated delayed diabetes progression, concurrent with reduced CD4+ T cell activation. Our results demonstrate that ROS are required for driving and sustaining T cell activation-induced metabolic reprogramming, and further support ROS as a target to minimize aberrant immune responses in autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Reactive Oxygen Species/metabolism , Aerobiosis , Animals , Cell Proliferation , Glucose/metabolism , Glycolysis , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , NADPH Oxidases/metabolism , Oxidative Phosphorylation
14.
Cell Microbiol ; 18(12): 1782-1799, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27206578

ABSTRACT

Borrelia burgdorferi possesses a sophisticated and complex chemotaxis system, but how the organism utilizes this system in its natural enzootic life cycle is poorly understood. Of the three CheY chemotaxis response regulators in B. burgdorferi, we found that only deletion of cheY3 resulted in an altered motility and significantly reduced chemotaxis phenotype. Although ΔcheY3 maintained normal densities in unfed ticks, their numbers were significantly reduced in fed ticks compared with the parental or cheY3-complemented spirochetes. Importantly, mice fed upon by the ΔcheY3-infected ticks did not develop a persistent infection. Intravital confocal microscopy analyses discovered that the ΔcheY3 spirochetes were motile within skin, but appeared unable to reverse direction and perform the characteristic backward-forward motility displayed by the parental strain. Subsequently, the ΔcheY3 became 'trapped' in the skin matrix within days of inoculation, were cleared from the skin needle-inoculation site within 96 h post-injection and did not disseminate to distant tissues. Interestingly, although ΔcheY3 cells were cleared within 96 h post-injection, this attenuated infection elicited significant levels of B. burgdorferi-specific IgM and IgG. Taken together, these data demonstrate that cheY3-mediated chemotaxis is crucial for motility, dissemination and viability of the spirochete both within and between mice and ticks.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Chemotaxis , Ixodes/microbiology , Lyme Disease/microbiology , Methyl-Accepting Chemotaxis Proteins/genetics , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Load , Bacterial Proteins/metabolism , Borrelia burgdorferi/growth & development , Borrelia burgdorferi/pathogenicity , Gene Deletion , Gene Expression , Genetic Complementation Test , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lyme Disease/immunology , Lyme Disease/pathology , Lyme Disease/transmission , Methyl-Accepting Chemotaxis Proteins/deficiency , Mice , Mice, Inbred C57BL , Phenotype , Skin/microbiology , Skin/pathology
15.
J Biol Chem ; 291(19): 10184-200, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26969166

ABSTRACT

Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) is the primary regulator of mitochondrial biogenesis and was recently found to be highly expressed within the intestinal epithelium. PGC1α is decreased in the intestinal epithelium of patients with inflammatory bowel disease, but its role in pathogenesis is uncertain. We now hypothesize that PGC1α protects against the development of colitis and helps to maintain the integrity of the intestinal barrier. We selectively deleted PGC1α from the intestinal epithelium of mice by breeding a PGC1α(loxP/loxP) mouse with a villin-cre mouse. Their progeny (PGC1α(ΔIEC) mice) were subjected to 2% dextran sodium sulfate (DSS) colitis for 7 days. The SIRT1 agonist SRT1720 was used to enhance PGC1α activation in wild-type mice during DSS exposure. Mice lacking PGC1α within the intestinal epithelium were more susceptible to DSS colitis than their wild-type littermates. Pharmacologic activation of PGC1α successfully ameliorated disease and restored mitochondrial integrity. These findings suggest that a depletion of PGC1α in the intestinal epithelium contributes to inflammatory changes through a failure of mitochondrial structure and function as well as a breakdown of the intestinal barrier, which leads to increased bacterial translocation. PGC1α induction helps to maintain mitochondrial integrity, enhance intestinal barrier function, and decrease inflammation.


Subject(s)
Colitis/metabolism , Intestinal Mucosa/metabolism , Mitochondria/metabolism , Transcription Factors/metabolism , Animals , Bacterial Translocation/drug effects , Bacterial Translocation/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics
16.
Front Cell Dev Biol ; 3: 62, 2015.
Article in English | MEDLINE | ID: mdl-26484345

ABSTRACT

Inflammatory Bowel Disease (IBD) represents a group of idiopathic disorders characterized by chronic or recurring inflammation of the gastrointestinal tract. While the exact etiology of disease is unknown, IBD is recognized to be a complex, multifactorial disease that results from an intricate interplay of genetic predisposition, an altered immune response, changes in the intestinal microbiota, and environmental factors. Together, these contribute to a destruction of the intestinal epithelial barrier, increased gut permeability, and an influx of immune cells. Given that most cellular functions as well as maintenance of the epithelial barrier is energy-dependent, it is logical to assume that mitochondrial dysfunction may play a key role in both the onset and recurrence of disease. Indeed several studies have demonstrated evidence of mitochondrial stress and alterations in mitochondrial function within the intestinal epithelium of patients with IBD and mice undergoing experimental colitis. Although the hallmarks of mitochondrial dysfunction, including oxidative stress and impaired ATP production are known to be evident in the intestines of patients with IBD, it is as yet unclear whether these processes occur as a cause of consequence of disease. We provide a current review of mitochondrial function in the setting of intestinal inflammation during IBD.

17.
Article in English | MEDLINE | ID: mdl-24822172

ABSTRACT

In nature, the Lyme disease spirochete Borrelia burgdorferi cycles between the unrelated environments of the Ixodes tick vector and mammalian host. In order to survive transmission between hosts, B. burgdorferi must be able to not only detect changes in its environment, but also rapidly and appropriately respond to these changes. One manner in which this obligate parasite regulates and adapts to its changing environment is through cyclic-di-GMP (c-di-GMP) signaling. c-di-GMP has been shown to be instrumental in orchestrating the adaptation of B. burgdorferi to the tick environment. B. burgdorferi possesses only one set of c-di-GMP-metabolizing genes (one diguanylate cyclase and two distinct phosphodiesterases) and one c-di-GMP-binding PilZ-domain protein designated as PlzA. While studies in the realm of c-di-GMP signaling in B. burgdorferi have exploded in the last few years, there are still many more questions than answers. Elucidation of the importance of c-di-GMP signaling to B. burgdorferi may lead to the identification of mechanisms that are critical for the survival of B. burgdorferi in the tick phase of the enzootic cycle as well as potentially delineate a role (if any) c-di-GMP may play in the transmission and virulence of B. burgdorferi during the enzootic cycle, thereby enabling the development of effective drugs for the prevention and/or treatment of Lyme disease.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/physiology , Cyclic GMP/analogs & derivatives , Lyme Disease/microbiology , Signal Transduction , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Animals , Carrier Proteins/metabolism , Cyclic GMP/metabolism , Escherichia coli Proteins/metabolism , Humans , Phosphorus-Oxygen Lyases/metabolism , Protein Binding
18.
Am J Physiol Gastrointest Liver Physiol ; 302(2): G250-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22094600

ABSTRACT

The n-3 fatty acids contribute to regulation of hepatic fatty acid oxidation and synthesis in adults and accumulate in fetal and infant liver in variable amounts depending on the maternal diet fat composition. Using 2D gel proteomics and matrix-assisted laser desorption/ionization time of flight mass spectrometry, we recently identified altered abundance of proteins associated with glucose and amino acid metabolism in neonatal rat liver with increased n-3 fatty acids. Here, we extend studies on n-3 fatty acids in hepatic metabolic development to targeted gene and metabolite analyses and map the results into metabolic pathways to consider the role of n-3 fatty acids in glucose, fatty acid, and amino metabolism. Feeding rats 1.5% compared with <0.1% energy 18:3n-3 during gestation led to higher 20:5n-3 and 22:6n-3 in 3-day-old offspring liver, higher serine hydroxymethyltransferase, carnitine palmitoyl transferase, and acyl CoA oxidase and lower pyruvate kinase and stearoyl CoA desaturase gene expression, with higher cholesterol, NADPH and glutathione, and lower glycine (P < 0.05). Integration of the results suggests that the n-3 fatty acids may be important in facilitating hepatic metabolic adaptation from in utero nutrition to the postnatal high-fat milk diet, by increasing fatty acid oxidation and directing glucose and amino acids to anabolic pathways.


Subject(s)
Fatty Acids, Omega-3/administration & dosage , Liver/enzymology , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cholesterol/metabolism , Gene Expression , Glutathione/genetics , Glutathione/metabolism , Glycine/genetics , Glycine/metabolism , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Lipid Metabolism , Liver/metabolism , NADP/genetics , NADP/metabolism , Oxidation-Reduction , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Rats , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
19.
Nestle Nutr Workshop Ser Pediatr Program ; 68: 17-27; discussion 27-32, 2011.
Article in English | MEDLINE | ID: mdl-22044889

ABSTRACT

Understanding the importance of dietary fat has grown beyond energy metabolism to recognition of the complex roles of fatty acids, particularly the ω-6 and ω-3 fatty acids in membrane lipids, inter- and intracellular communication and in regulating gene expression. The ω-6 and ω-3 fatty acids accumulated in developing tissues depend on the fatty acids transported across the placenta and secreted in breast milk. These in turn are dependent on maternal fatty acid intakes, which have changed dramatically in the past century with current western diets high in ω-6 linoleic acid and low in ω-3 fatty acids. High intakes of ω-6 fatty acid and low intakes of ω-3 fatty acids compromise long-chain ω-3 fatty acid accumulation in tissues, and this is avoided by dietary docosahexaenoic acid. In addition to the well-known roles in neural development, newer studies are beginning to question the importance of ω-3 fatty acids as a contributor of metabolic development in other organs, with possible implications for the development of feeding behavior and integration of the nutrient energy supply.


Subject(s)
Dietary Fats , Health Status , Adult , Animals , Breast Feeding , Child Development , Dietary Fats/adverse effects , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/administration & dosage , Female , Fetal Development , Humans , Infant , Infant, Newborn , Lipid Metabolism , Male , Maternal Nutritional Physiological Phenomena , Pregnancy
20.
Am J Physiol Endocrinol Metab ; 301(5): E807-17, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21791621

ABSTRACT

Levels of n-6, n-3, and medium-chain fatty acids (MCFA) in milk are highly variable. Higher carbohydrate intakes are associated with increased mammary gland MCFA synthesis, but the role of unsaturated fatty acids for milk MCFA secretion is unclear. This study addressed whether n-6 and n-3 fatty acids, which are known to inhibit hepatic fatty acid synthesis, influence MCFA in rat and human milk and the implications of varying MCFA, n-6, and n-3 fatty acids in rat milk for metabolic regulation in the neonatal liver. Rats were fed a low-fat diet or one of six higher-fat diets, varying in 16:0, 18:1n-9, 18:2n-6, 18:3n-3, and long-chain (LC) n-3 fatty acids. Higher maternal dietary 18:2n-6 or 18:3n-3 did not influence milk MCFA, but lower maternal plasma triglycerides, due to either a low-fat or a high-fat high-LC n-3 diet led to higher milk MCFA. MCFA levels were inversely associated with 18:1n-9, 18:2n-6, and 18:3n-3 in human milk, likely reflecting the association between dietary total fat and unsaturated fatty acids. High LC n-3 fatty acid in rat milk was associated with lower hepatic Pklr, Acly, Fasn, and Scd1 and higher Hmgcs2 in the milk-fed rat neonate, with no effect of milk 18:1n-9, 18:2n-6, or MCFA. These studies show that the dietary fatty acid composition does not impact MCFA secretion in milk, but the fatty acid composition of milk, particularly the LC n-3 fatty acid, is relevant to hepatic metabolic regulation in the milk-fed neonate.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/pharmacology , Fatty Acids/metabolism , Liver/drug effects , Maternal Nutritional Physiological Phenomena , Milk/drug effects , Animals , Animals, Newborn , Dietary Fats/pharmacology , Fatty Acids/analysis , Female , Liver/metabolism , Liver/physiology , Maternal Nutritional Physiological Phenomena/drug effects , Milk/chemistry , Milk/metabolism , Parturition/drug effects , Parturition/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...