Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Genome Biol ; 17(1): 201, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27687735

ABSTRACT

We present a sensitive approach to predict genes expressed selectively in specific cell types, by searching publicly available expression data for genes with a similar expression profile to known cell-specific markers. Our method, CellMapper, strongly outperforms previous computational algorithms to predict cell type-specific expression, especially for rare and difficult-to-isolate cell types. Furthermore, CellMapper makes accurate predictions for human brain cell types that have never been isolated, and can be rapidly applied to diverse cell types from many tissues. We demonstrate a clinically relevant application to prioritize candidate genes in disease susceptibility loci identified by GWAS.


Subject(s)
Algorithms , Computational Biology/methods , Gene Expression Profiling/methods , Genome-Wide Association Study/methods , Animals , Brain/cytology , Brain/metabolism , Caco-2 Cells , Cell Line , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genetic Predisposition to Disease/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immune System/cytology , Immune System/metabolism , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Polymorphism, Single Nucleotide , Reproducibility of Results , Spleen/cytology , Spleen/metabolism
2.
Cell Rep ; 13(11): 2403-2411, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26686631

ABSTRACT

The cellular and molecular mechanisms underlying adaptive changes to physiological stress within the intestinal epithelium remain poorly understood. Here, we show that PTEN, a negative regulator of the PI3K→AKT→mTORC1-signaling pathway, is an important regulator of dormant intestinal stem cells (d-ISCs). Acute nutrient deprivation leads to transient PTEN phosphorylation within d-ISCs and a corresponding increase in their number. This release of PTEN inhibition renders d-ISCs functionally poised to contribute to the regenerative response during re-feeding via cell-autonomous activation of the PI3K→AKT→mTORC1 pathway. Consistent with its role in mediating cell survival, PTEN is required for d-ISC maintenance at baseline, and intestines lacking PTEN have diminished regenerative capacity after irradiation. Our results highlight a PTEN-dependent mechanism for d-ISC maintenance and further demonstrate the role of d-ISCs in the intestinal response to stress.


Subject(s)
Intestines/cytology , Nutritional Status , PTEN Phosphohydrolase/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , Female , Genes, Reporter , Intestines/pathology , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Telomerase/genetics , Telomerase/metabolism
3.
J Pediatr Gastroenterol Nutr ; 60(2): 182-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25625576

ABSTRACT

OBJECTIVES: Recent data from mainly homogeneous European and African populations implicate a 140-bp region 5' to the transcriptional start site of LCT (the lactase gene) as a regulatory site for lactase persistence and nonpersistence. Because there are no studies of US nonhomogeneous populations, we performed genotype/phenotype analysis of the -13910 and -22018 LCT single nucleotide polymorphisms (SNPs) in New England children, mostly of European ancestry. METHODS: Duodenal biopsies were processed for disaccharidase activities, RNA quantification by reverse transcription polymerase chain reaction (RT-PCR), allelic expression ratios by PCR, and genotyping and SNP analysis. Results were compared with clinical information. RESULTS: Lactase activity and mRNA levels, and sucrase-to-lactase ratios of enzyme activity and mRNA, showed robust correlations with genotype. None of the other LCT SNPs showed as strong a correlation with enzyme or mRNA levels as did -13910. Data were consistent, with the -13910 being the causal sequence variant instead of -22018. Four individuals heterozygous for -13910T/C had allelic expression patterns similar to individuals with -13910C/C genotypes; of these, 2 showed equal LCT expression from the 2 alleles and a novel variant (-13909C>A) associated with lactase persistence. CONCLUSIONS: The identification of -13910C/C genotype is likely to predict lactase nonpersistence, consistent with prior published studies. A -13910T/T genotype will frequently, but not perfectly, predict lactase persistence in this mixed European-ancestry population; a -13910T/C genotype will not predict the phenotype. A long, rare haplotype in 2 individuals with -13910T/C genotype but equal allele-specific expression contains a novel lactase persistence allele present at -13909.


Subject(s)
Duodenum/enzymology , Lactase/genetics , Lactase/metabolism , RNA, Messenger/metabolism , White People/genetics , Adolescent , Alleles , Child , Duodenum/metabolism , Female , Genotype , Humans , Male , Phenotype , Polymorphism, Single Nucleotide , Sucrase/metabolism , United States/ethnology , Young Adult
4.
Dig Dis Sci ; 56(1): 59-69, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21086165

ABSTRACT

BACKGROUND AND AIM: The regulation of human intestinal lactase-phlorizin hydrolase remains incompletely understood. One kb of pig and 2 kb of rat 5'-flanking sequence controls correct tissue, cell, topographic, and villus LCT expression. To gain insight into human LCT expression, transgenic mouse lines were generated from 3.3 kb of human LPH 5' flanking sequence from a lactase persistent individual fused to a human growth hormone (hGH) reporter bounded by an insulator. METHODS: Four lines were identified in which reporter expression was specifically detectable in the intestine and no other organ, two of which demonstrated hGH expression specific to small and large intestine. Quantitative RT-PCR was carried out on proximal to distal segments of small intestine at fetal days 16.5 and 18.5 and at birth, postnatal days 7 and 28 in line 22. RESULTS: In fetal intestine, hGH expression demonstrated a proximal to distal gradient similar to that in native intestine. There was no significant difference between hGH expression levels at 7 and 28 days in segment 3, the midpoint of the small intestine, where expression of endogenous lactase is maximal at 7 days and declines significantly by 28 days. Distal small intestine displayed high levels of hGH expression in enteroendocrine cells, which were shown to be a subset of the PYY cells. CONCLUSIONS: Thus, a 3.3-kb LPH 5' flanking sequence construct from a lactase persistent individual is able to maintain postnatal expression in transgenic mice post weaning.


Subject(s)
5' Flanking Region/genetics , Intestine, Small/enzymology , Lactase-Phlorizin Hydrolase/genetics , Lactase-Phlorizin Hydrolase/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Epithelial Cells/cytology , Epithelial Cells/enzymology , Female , Fetus/enzymology , Growth Hormone/genetics , Growth Hormone/metabolism , Humans , Intestine, Small/cytology , Intestine, Small/embryology , Male , Mice , Mice, Transgenic
5.
Proc Natl Acad Sci U S A ; 108(1): 179-84, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21173232

ABSTRACT

The intestinal epithelium is maintained by a population of rapidly cycling (Lgr5(+)) intestinal stem cells (ISCs). It has been postulated, however, that slowly cycling ISCs must also be present in the intestine to protect the genome from accumulating deleterious mutations and to allow for a response to tissue injury. Here, we identify a subpopulation of slowly cycling ISCs marked by mouse telomerase reverse transcriptase (mTert) expression that can give rise to Lgr5(+) cells. mTert-expressing cells distribute in a pattern along the crypt-villus axis similar to long-term label-retaining cells (LRCs) and are resistant to tissue injury. Lineage-tracing studies demonstrate that mTert(+) cells give rise to all differentiated intestinal cell types, persist long term, and contribute to the regenerative response following injury. Consistent with other highly regenerative tissues, our results demonstrate that a slowly cycling stem cell population exists within the intestine.


Subject(s)
Intestinal Mucosa/cytology , Multipotent Stem Cells/metabolism , Telomerase/metabolism , Animals , Cell Lineage/physiology , Flow Cytometry , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice , Microscopy, Fluorescence , Multipotent Stem Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Dev Cell ; 19(5): 713-26, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21074721

ABSTRACT

VIDEO ABSTRACT: Cell differentiation requires remodeling of tissue-specific gene loci and activities of key transcriptional regulators, which are recognized for their dominant control over cellular programs. Using epigenomic methods, we characterized enhancer elements specifically modified in differentiating intestinal epithelial cells and found enrichment of transcription factor-binding motifs corresponding to CDX2, a critical regulator of the intestine. Directed investigation revealed surprising lability in CDX2 occupancy of the genome, with redistribution from hundreds of sites occupied only in proliferating cells to thousands of new sites in differentiated cells. Knockout mice confirmed distinct Cdx2 requirements in dividing and mature adult intestinal cells, including responsibility for the active enhancer configuration associated with maturity. Dynamic CDX2 occupancy corresponds with condition-specific gene expression and, importantly, to differential co-occupancy with other tissue-restricted transcription factors, such as GATA6 and HNF4A. These results reveal dynamic, context-specific functions and mechanisms of a prominent transcriptional regulator within a cell lineage.


Subject(s)
Cell Differentiation/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Homeodomain Proteins/metabolism , Intestines/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , CDX2 Transcription Factor , Caco-2 Cells , Cell Proliferation , Chromatin/chemistry , Chromatin/genetics , Enhancer Elements, Genetic , Epigenomics/methods , Epithelial Cells/cytology , Epithelial Cells/physiology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Genome , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Histones/genetics , Homeodomain Proteins/genetics , Humans , Intestines/cytology , Mice , Mice, Knockout , Molecular Sequence Data , Trans-Activators/genetics , Transcription Factors/genetics
7.
Dig Dis Sci ; 55(9): 2478-88, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20112066

ABSTRACT

BACKGROUND: Eph receptor tyrosine kinases EphB2 and EphB3, and ephrin-B1 ligand play a critical role in regulating small intestinal epithelial cell migration. Although well studied in developing brain, the expression pattern of Ephs/ephrins has not been delineated in the developing small intestine. AIMS: To examine the gene expression of all known members of Ephs/ephrins during development of mouse small intestine. METHODS: We examined the expression of 21 A- and B-Ephs/ephrins in mouse small intestine or the Caco-2 cell line using reverse-transcription polymerase chain reaction (RT-PCR), quantitative (q)RT-PCR, and immunohistochemical analyses. EphB2-expressing cells from isolated crypts were detected by immunofluorescence and fluorescence-activated cell sorting (FACS) analyses. RESULTS: With the exception of EphA5, all family members were expressed throughout the intestine at all ages examined. Most were uniformly expressed. In contrast, levels of EphA4, EphA8, EphB4, and ephrin-B2 messenger RNA (mRNA) were highest during early fetal development and declined with age. At E15, EphB2 and EphB4 proteins were diffusely expressed in proliferating stratified intestinal epithelial cells. By E18, the proteins had become localized to cell membranes of columnar epithelial cells within intervillus regions, and later were expressed on epithelial cell membranes in adult crypts. EphB2-expressing cells can be specifically isolated from crypt cell fractions. CONCLUSIONS: The current study represents the first analysis of Ephs/ephrins during intestinal development. The elevated expression of EphA4, EphA8, EphB4, and ephrin-B2 during the fetal period of intestinal morphogenesis suggests an important role in development. Continued intestinal expression of other family members implicates a role in differentiation.


Subject(s)
Ephrins/metabolism , Intestine, Small/metabolism , Receptors, Eph Family/metabolism , Age Factors , Animals , Caco-2 Cells , Cell Differentiation , Cell Separation , Ephrins/genetics , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Gestational Age , Humans , Immunohistochemistry , Intestine, Small/embryology , Intestine, Small/growth & development , Mice , Mice, Inbred C57BL , Morphogenesis , RNA, Messenger/metabolism , Receptors, Eph Family/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
J Anat ; 213(1): 52-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18638070

ABSTRACT

Stem cells hold great promise for regenerative medicine but remain elusive in many tissues, including the small intestine, where it is well accepted that the epithelium is maintained by intestinal stem cells located in the crypts. The lack of established markers to prospectively identify intestinal stem cells has necessitated the use of indirect analysis, e.g. long-term label retention, which is based on the hypothesis that intestinal stem cells are slow-cycling. Several intestinal stem cell markers have been proposed, including Musashi-1, BMPR1alpha, phospho-PTEN, DCAMKL1, Eph receptors and integrins, but their validity, using functional and/or lineage tracing assays, has yet to be confirmed. Recently, Lgr5 has been identified by lineage tracing as an intestinal stem cell marker. In this review we summarize what is known about the currently reported intestinal stem cell markers and provide a rationale for developing model systems whereby intestinal stem cells can be functionally validated.


Subject(s)
Intestine, Small/metabolism , Stem Cells/metabolism , Biomarkers/analysis , Cell Lineage , Humans , Intestine, Small/cytology , Research
10.
Proc Natl Acad Sci U S A ; 105(30): 10420-5, 2008 Jul 29.
Article in English | MEDLINE | ID: mdl-18650388

ABSTRACT

Stem cells hold great promise for regenerative medicine, but remain elusive in many tissues in part because universal markers of "stemness" have not been identified. The ribonucleoprotein complex telomerase catalyzes the extension of chromosome ends, and its expression is associated with failure of cells to undergo cellular senescence. Because such resistance to senescence is a common characteristic of many stem cells, we hypothesized that telomerase expression may provide a selective biomarker for stem cells in multiple tissues. In fact, telomerase expression has been demonstrated within hematopoietic stem cells. We therefore generated mouse telomerase reverse transcriptase (mTert)-GFP-transgenic mice and assayed the ability of mTert-driven GFP to mark tissue stem cells in testis, bone marrow (BM), and intestine. mTert-GFP mice were generated by using a two-step embryonic stem cell-based strategy, which enabled primary and secondary screening of stably transfected clones before blastocyst injection, greatly increasing the probability of obtaining mTert reporter mice with physiologically appropriate regulation of GFP expression. Analysis of adult mice showed that GFP is expressed in differentiating male germ cells, is enriched among BM-derived hematopoietic stem cells, and specifically marks long-term BrdU-retaining intestinal crypt cells. In addition, telomerase-expressing GFP(+) BM cells showed long-term, serial, multilineage BM reconstitution, fulfilling the functional definition of hematopoietic stem cells. Together, these data provide direct evidence that mTert-GFP expression marks progenitor cells in blood and small intestine, validating these mice as a useful tool for the prospective identification, isolation, and functional characterization of progenitor/stem cells from multiple tissues.


Subject(s)
Embryonic Stem Cells/cytology , Genetic Techniques , Green Fluorescent Proteins/metabolism , Stem Cells/cytology , Telomerase/metabolism , Animals , Biomarkers/metabolism , Bone Marrow/metabolism , Cell Separation , Intestinal Mucosa/metabolism , Male , Mice , Mice, Transgenic , Phenotype , Testis/metabolism
11.
Curr Treat Options Gastroenterol ; 11(1): 19-25, 2008 Feb.
Article in English | MEDLINE | ID: mdl-21063860

ABSTRACT

OPINION STATEMENT: Lactose malabsorption is a syndrome producing constellation of symptoms, including abdominal pain, bloating, flatulence, diarrhea, and sometimes nausea and/or vomiting. Primary causes of lactose malabsorption due to loss of intestinal lactase activity include genetic/racial lactase nonpersistence, congenital lactase deficiency, and developmental lactase deficiency. Secondary lactose malabsorption can be caused by any disorder that injures the small intestinal mucosa, such as viral gastroenteritis, celiac disease, allergic (eosinophilic) gastroenteritis, and radiation enteritis. The diagnosis depends on careful clinical evaluation and is customarily confirmed with a lactose breath hydrogen test. As the symptoms are nonspecific, many adults diagnosed with lactose malabsorption actually have irritable bowel syndrome. Treatment consists of a trial of eliminating lactose-containing dairy foods, with supplementation of alternative calcium and protein sources. Commercial enzyme products containing ß-galactosidases can be prescribed to help patients digest dietary lactose. Long-term lactose restriction usually is not necessary and can lead to reduced bone mineral density.

12.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1302-14, 2007 May.
Article in English | MEDLINE | ID: mdl-17272516

ABSTRACT

The terminal differentiation phases of intestinal development in mice occur during cytodifferentiation and the weaning transition. Lactase-phlorizin hydrolase (LPH), liver fatty acid binding protein (Fabp1), and sucrase-isomaltase (SI) are well-characterized markers of these transitions. With the use of gene inactivation models in mature mouse jejunum, we have previously shown that a member of the zinc finger transcription factor family (Gata4) and hepatocyte nuclear factor-1alpha (Hnf1alpha) are each indispensable for LPH and Fabp1 gene expression but are both dispensable for SI gene expression. In the present study, we used these models to test the hypothesis that Gata4 and Hnf1alpha regulate LPH, Fabp1, and SI gene expression during development, specifically focusing on cytodifferentiation and the weaning transition. Inactivation of Gata4 had no effect on LPH gene expression during either cytodifferentiation or suckling, whereas inactivation of Hnf1alpha resulted in a 50% reduction in LPH gene expression during these same time intervals. Inactivation of Gata4 or Hnf1alpha had a partial effect ( approximately 50% reduction) on Fabp1 gene expression during cytodifferentiation and suckling but no effect on SI gene expression at any time during development. Throughout the suckling period, we found a surprising and dramatic reduction in Gata4 and Hnf1alpha protein in the nuclei of absorptive enterocytes of the jejunum despite high levels of their mRNAs. Finally, we show that neither Gata4 nor Hnf1alpha mediates the glucocorticoid-induced precocious maturation of the intestine but rather are downstream targets of this process. Together, these data demonstrate that specific intestinal genes have differential requirements for Gata4 and Hnf1alpha that are dependent on the developmental time frame in which they are expressed.


Subject(s)
Fatty Acid-Binding Proteins/biosynthesis , GATA4 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/physiology , Intestine, Small/growth & development , Lactase-Phlorizin Hydrolase/biosynthesis , Sucrase-Isomaltase Complex/biosynthesis , Animals , Female , Glucocorticoids/pharmacology , Intestine, Small/drug effects , Intestine, Small/embryology , Mice , Pregnancy , RNA, Messenger/metabolism , Weaning
13.
J Pediatr Gastroenterol Nutr ; 45 Suppl 2: S131-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18185074

ABSTRACT

Lactase-phlorizin hydrolase (LPH) is expressed only in the small intestine and is confined to absorptive enterocytes on the villi with a tightly controlled pattern of expression along the proximal to distal and crypt-villus axes of the intestine. LPH expression is regulated mainly at the level of lactase (LCT) gene transcription that directs 2 phenotypes: a decline in LCT activity (LCT nonpersistence) in mid-childhood in the majority of the world's population, and maintenance of the lactase levels found in infancy (LCT persistence) in people of northern European extraction and scattered populations elsewhere. The molecular mechanisms that regulate these phenotypes are not completely understood. A population genetic association of lactase persistence with 2 single nucleotide polymorphisms in the distal 5'-flanking region of LCT (-13.9T and -22A) has been confirmed in northern Europeans, but this fails to explain lactase persistence found in some African groups. Any hypothesis for the control of lactase expression must reconcile the presence of high levels of activity in early life in all humans and the characteristic loss of activity found subsequently in many but not all people.


Subject(s)
Gene Expression Regulation, Enzymologic , Lactase-Phlorizin Hydrolase/metabolism , Lactase/genetics , Lactose/metabolism , Polymorphism, Single Nucleotide , Genetics, Population , Genotype , Humans , Intestine, Small/enzymology
14.
Am J Physiol Gastrointest Liver Physiol ; 290(5): G1016-24, 2006 May.
Article in English | MEDLINE | ID: mdl-16223943

ABSTRACT

Hepatocyte nuclear factor-1alpha (HNF-1alpha) is a modified homeodomain-containing transcription factor that has been implicated in the regulation of intestinal genes. To define the importance and underlying mechanism of HNF-1alpha for the regulation of intestinal gene expression in vivo, we analyzed the expression of the intestinal differentiation markers and putative HNF-1alpha targets lactase-phlorizin hydrolase (LPH) and sucrase-isomaltase (SI) in hnf1alpha null mice. We found that in adult jejunum, LPH mRNA in hnf1alpha(-/-) mice was reduced 95% compared with wild-type controls (P < 0.01, n = 4), whereas SI mRNA was virtually identical to that in wild-type mice. Furthermore, SI mRNA abundance was unchanged in the absence of HNF-1alpha along the length of the adult mouse small intestine as well as in newborn jejunum. We found that HNF-1alpha occupies the promoters of both the LPH and SI genes in vivo. However, in contrast to liver and pancreas, where HNF-1alpha regulates target genes by recruitment of histone acetyl transferase activity to the promoter, the histone acetylation state of the LPH and SI promoters was not affected by the presence or absence of HNF-1alpha. Finally, we showed that a subset of hypothesized intestinal target genes is regulated by HNF-1alpha in vivo and that this regulation occurs in a defined tissue-specific and developmental context. These data indicate that HNF-1alpha is an activator of a subset of intestinal genes and induces these genes through an alternative mechanism in which it is dispensable for chromatin remodeling.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 1/metabolism , Histones/metabolism , Lactase-Phlorizin Hydrolase/genetics , Lactase-Phlorizin Hydrolase/metabolism , Acetylation , Animals , Genes, Reporter , Hepatocyte Nuclear Factor 1/genetics , Hepatocyte Nuclear Factor 1/physiology , Intestinal Mucosa/metabolism , Jejunum/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Sucrase-Isomaltase Complex/metabolism , Transcription Factors/metabolism
15.
J Histochem Cytochem ; 53(12): 1501-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16009965

ABSTRACT

RNA localization is a regulated component of gene expression of fundamental importance in development and differentiation. Several RNA binding proteins involved in RNA localization during development in Drosophila have been identified, of which Y14, Mago, Pumilio, and IMP-1 are known to be expressed in adult mammalian intestine. The present study was undertaken to define the developmental and regional expression of these proteins, as well as Staufen-1, in mouse intestinal cells and in other tissues and cell lines using RT-PCR, and localization using in situ hybridization and immunohistochemistry. Staufen-1, Y14, Mago-m, and Pumilio-1 were expressed in intestinal epithelial cells of both villus and crypt and in Caco-2 and IEC-6 cells. In contrast, expression of IMP-1 was age- and region-specific, showing clear expression in distal fetal and newborn intestine, but very low or no expression in adult. The mRNAs were cytosolic, with more apical than basal expression in enterocytes. Staufen protein showed a similar localization pattern to that of its cognate mRNA. Overall, the data suggest an essential role for these proteins in intestinal cells. Age and regional expression of IMP-1 may indicate a role in regulation of site-specific translation of intestinal genes or in RNA localization.


Subject(s)
Intestinal Mucosa/metabolism , RNA Transport , RNA, Messenger/biosynthesis , RNA-Binding Proteins/biosynthesis , Animals , Animals, Newborn , Cell Line , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , In Situ Hybridization , Intestinal Mucosa/embryology , Intestinal Mucosa/growth & development , Mice , Organ Specificity , RNA-Binding Proteins/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction
16.
J Pediatr Gastroenterol Nutr ; 39(3): 275-85, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15319629

ABSTRACT

OBJECTIVES: Lactase-phlorizin hydrolase (LPH) is an enterocyte-specific gene whose expression has been well-characterized, not only developmentally but also along the crypt-villus axis and along the length of the small bowel. Previous studies from the authors' laboratory have demonstrated that 2 kb of the 5'-flanking region of the rat LPH gene control the correct tissue, cell, and crypt-villus expression in transgenic animals. METHODS: To examine further the regulation conferred by this region, protein-DNA interactions were studied using DNase I footprint analyses in LPH-expressing and nonexpressing cell lines. Functional delineation of this 5'-flanking sequence was performed using deletion analysis in transient transfection assays. RESULTS: Studies revealed a generally positive activity between -74 and -37 bp, a cell-specific negative region between -210 and -95 bp, and additional elements further toward the 5'-terminus that conferred a highly cell-specific response in reporter activity. Computer analysis of distal regions encompassing identified footprints revealed potential binding sites for various intestinal transcription factors. Co-transfection and electromobility shift assay experiments indicated binding of HNF3beta at three sites relevant to LPH expression. CONCLUSIONS: The data demonstrate that the cell specificity of LPH gene expression depends upon both positive and negative interactions among elements in the first 2 kb of the LPH 5'-flanking region.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Enzymologic , Lactase-Phlorizin Hydrolase/metabolism , Transcription Factors/physiology , Animals , Caco-2 Cells , DNA Footprinting , Electrophoretic Mobility Shift Assay , Humans , Lactase-Phlorizin Hydrolase/genetics , Molecular Weight , Rats , Transcription, Genetic , Transfection
17.
Am J Physiol Gastrointest Liver Physiol ; 287(4): G899-909, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15178553

ABSTRACT

Lactase-phlorizin hydrolase (LPH), a marker of intestinal differentiation, is expressed in absorptive enterocytes on small intestinal villi in a tightly regulated pattern along the proximal-distal axis. The LPH promoter contains binding sites that mediate activation by members of the GATA-4, -5, and -6 subfamily, but little is known about their individual contribution to LPH regulation in vivo. Here, we show that GATA-4 is the principal GATA factor from adult mouse intestinal epithelial cells that binds to the mouse LPH promoter, and its expression is highly correlated with that of LPH mRNA in jejunum and ileum. GATA-4 cooperates with hepatocyte nuclear factor (HNF)-1alpha to synergistically activate the LPH promoter by a mechanism identical to that previously characterized for GATA-5/HNF-1alpha, requiring physical association between GATA-4 and HNF-1alpha and intact HNF-1 binding sites on the LPH promoter. GATA-4 also activates the LPH promoter independently of HNF-1alpha, in contrast to GATA-5, which is unable to activate the LPH promoter in the absence of HNF-1alpha. GATA-4-specific activation requires intact GATA binding sites on the LPH promoter and was mapped by domain-swapping experiments to the zinc finger and basic regions. However, the difference in the capacity between GATA-4 and GATA-5 to activate the LPH promoter was not due to a difference in affinity for binding to GATA binding sites on the LPH promoter. These data indicate that GATA-4 is a key regulator of LPH gene expression that may function through an evolutionarily conserved mechanism involving cooperativity with an HNF-1alpha and/or a GATA-specific pathway independent of HNF-1alpha.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Intestinal Mucosa/physiology , Lactase-Phlorizin Hydrolase/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Cell Differentiation/physiology , GATA4 Transcription Factor , Genes, Reporter , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Humans , Intestinal Mucosa/cytology , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Transfection , Zinc Fingers/physiology
18.
Crit Care Nurs Clin North Am ; 16(1): 39-49, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15062412

ABSTRACT

Traumatic bum injuries and the associated treatments are a tremendous pain management challenge. The degree of tissue damage in severe burns can initiate physiologic changes in nociceptive pathways that place the patient at risk for undertreatment. The use of analgesic guidelines that address both background and procedural pain and associated anxiety can provide a rational and consistent approach to treatment. The key to successful treatment is the continuous and accurate assessment of the patient's pain and the response to therapy. Medications, especially opioids, should be regularly evaluated and adjusted to achieve maximum effect and minimal side effect.Nursing's role is perhaps the most important in the essential focused surveillance of bum pain and it's successful treatment.


Subject(s)
Burns/complications , Critical Care/methods , Pain , Analgesia/methods , Analgesia/nursing , Analgesics/therapeutic use , Anxiety/etiology , Anxiety/prevention & control , Critical Care/standards , Humans , Nurse's Role , Nursing Assessment/methods , Pain/diagnosis , Pain/drug therapy , Pain/etiology , Pain Measurement/methods , Practice Guidelines as Topic , Risk Factors
19.
Am J Physiol Gastrointest Liver Physiol ; 284(4): G722-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12490432

ABSTRACT

An important feature of enterocyte maturation is the asymmetrical distribution of cellular functions including protein localization. mRNA sorting is one mechanism for establishment and maintenance of this process in other systems, and we have previously demonstrated differential localization of mRNAs in human enterocytes. To study regulation of mRNA sorting, we established a model in polarized Caco-2 cells. Proxy cDNA constructs containing beta-galactosidase (beta-gal)/green fluorescence protein (GFP) and the 3'-untranslated region (3'-UTR) of either human sucrase-isomaltase or villin were transfected transiently or stably. A control construct contained poly-A sequence in place of 3'-UTR. Expression of GFP was observed by confocal microscopy; intracellular location of the construct mRNA was imaged by in situ hybridization. The sucrase-isomaltase mRNA proxy localized to an apical position in Caco-2 cells as in native enterocytes; the villin mRNA proxy did not show significant localization. The control construct was not localized and was found diffusely throughout the cell. Proxy GFP proteins tended to localize with their mRNA proxies, but with less precision. This study establishes a valuable model for the investigation of mRNA localization in intestinal epithelial cells. Mechanisms controlling asymmetrical distribution of intestinal mRNAs can be now be elucidated.


Subject(s)
Enterocytes/cytology , Enterocytes/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Sucrase-Isomaltase Complex/genetics , 3' Untranslated Regions/genetics , Caco-2 Cells , Carrier Proteins/genetics , Cell Polarity/physiology , Gene Expression/physiology , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , Microfilament Proteins/genetics , RNA, Messenger/analysis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...