Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neurogastroenterol Motil ; 23(2): 131-8, e26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20939847

ABSTRACT

BACKGROUND: Gastrointestinal dysfunction is very common in diabetic patients. We assessed the changes in the colonic enteric nervous system using colectomy specimens and intestinal biopsies from diabetic subjects and age-matched controls. METHODS: In control and diabetic colons, we determined the total ganglion area (hematoxylin-eosin staining), changes in neuronal markers-protein gene product 9.5, peripherin, neuronal nitric oxide synthase (nNOS), neuropeptide Y (NPY), choline acetyl transferase (ChAT) and vasoactive intestinal peptide (by immunostaining), apoptosis (cleaved caspase-3 staining) and reduced glutathione levels. Superoxide dismutase mRNA was determined in enteric ganglia isolated by laser capture micro dissection. Isometric muscle recording was used to assess contraction and relaxation responses of colonic circular muscle strips. Apoptosis in enteric neurons under hyperglycemia in vitro was determined by cleaved caspase-3 Western blotting and protective effects of lipoic acid were evaluated. KEY RESULTS: Diabetic subjects had higher incidence of lower gastrointestinal symptoms like constipation and diarrhea at baseline prior to surgery. Diabetic ganglia displayed significant decrease in ganglion size due to enhanced apoptosis and loss of peripherin, nNOS, NPY, and ChAT neurons. Reduced glutathione levels in the diabetic colon (HbA1C > 7%) were significantly less than the control, indicating increased oxidative stress. Colonic circular muscle strips from diabetic subjects showed impaired contraction and relaxation responses compared with the healthy controls. Hyperglycemia-induced cleaved caspase-3 in enteric neurons was reversed by lipoic acid. CONCLUSIONS & INFERENCES: Our data demonstrate loss of enteric neurons in the colon due to increased oxidative stress and apoptosis which may cause the motility disturbances seen in human diabetes. Antioxidants may be of therapeutic value for preventing motility disorders in diabetes.


Subject(s)
Apoptosis , Colon/innervation , Colon/physiopathology , Diabetes Complications/complications , Enteric Nervous System/pathology , Gastrointestinal Diseases/etiology , Oxidative Stress/physiology , Aged , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Biopsy , Case-Control Studies , Cell Line , Disease Models, Animal , Electric Stimulation , Enteric Nervous System/drug effects , Enteric Nervous System/physiopathology , Female , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/physiopathology , Humans , Male , Mice , Middle Aged , Muscle Contraction/physiology , Muscle Relaxation/physiology , Phosphatidylinositol 3-Kinases/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Thioctic Acid/pharmacology
2.
Biochim Biophys Acta ; 1769(2): 106-16, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17321610

ABSTRACT

Intestinal epithelial cells respond to inflammatory extracellular stimuli by activating mitogen activated protein kinase (MAPK) signaling, which mediates numerous pathophysiological effects, including intestinal inflammation. Here, we show that a novel isoform of SPS1-related proline alanine-rich kinase (SPAK/STE20) is involved in this inflammatory signaling cascade. We cloned and characterized a SPAK isoform from inflamed colon tissue, and found that this SPAK isoform lacked the characteristic PAPA box and alphaF loop found in SPAK. Based on genomic sequence analysis the lack of PAPA box and alphaF loop in colonic SPAK isoform was the result of specific splicing that affect exon 1 and exon 7 of the SPAK gene. The SPAK isoform was found in inflamed and non-inflamed colon tissues as well as Caco2-BBE cells, but not in other tissues, such as liver, spleen, brain, prostate and kidney. In vitro analyses demonstrated that the SPAK isoform possessed serine/threonine kinase activity, which could be abolished by a substitution of isoleucine for the lysine at position 34 in the ATP-binding site of the catalytic domain. Treatment of Caco2-BBE cells with the pro-inflammatory cytokine, interferon gamma, induced expression of the SPAK isoform. Over-expression of the SPAK isoform in Caco2-BBE cells led to nuclear translocation of an N-terminal fragment of the SPAK isoform, as well as activation of p38 MAP kinase signaling cascades and increased intestinal barrier permeability. These findings collectively suggest that pro-inflammatory cytokine signaling may induce expression of this novel SPAK isoform in intestinal epithelia, triggering the signaling cascades that govern intestinal inflammation.


Subject(s)
Colitis/enzymology , Intestinal Mucosa/enzymology , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Base Sequence , Caco-2 Cells , Cloning, Molecular , Colitis/genetics , Colon/enzymology , Humans , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Neuroscience ; 143(1): 241-51, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16996218

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) promotes the growth and survival of enteric neurons, but the mechanisms involved are poorly understood. GDNF is known to promote the survival of enteric neurons through activation of the PI3-Kinase/Akt signaling pathway. We investigated the role of glycogen synthase kinase-3beta (GSK-3beta) in enteric neuronal survival, and the ability of GDNF to regulate the activity of GSK-3beta using primary rat embryonic enteric neurons. GDNF, through activation of the PI3-kinase pathway enhanced the phosphorylation of GSK-3beta at its N-terminal serine-9 residue, and promoted the association of GSK-3beta with 14-3-3. Transfection of a constitutively active S9A-GSK-3beta mutant prevented the survival effects of GDNF, whereas a dominant negative GSK-3beta construct prevented GDNF withdrawal-induced cell death. Increased GSK-3beta activity was associated with an increase in tau phosphorylation. Thus, GDNF promotes enteric neuronal survival by modulating GSK-3beta and its downstream target tau. Inhibitors of GSK-3beta activity may have therapeutic potential in improving enteric neuronal survival.


Subject(s)
14-3-3 Proteins/metabolism , Enteric Nervous System/cytology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glycogen Synthase Kinase 3/metabolism , Neurons/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Cell Survival/physiology , Cells, Cultured , Chromones/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Embryo, Nonmammalian , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta , Immunohistochemistry/methods , Morpholines/pharmacology , Mutagenesis/physiology , Neurons/metabolism , Neurons/physiology , Phosphorylation/drug effects , Rats , Transfection/methods , Xenopus , Xenopus Proteins/metabolism
4.
Cell Mol Life Sci ; 62(22): 2647-57, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16322943

ABSTRACT

Adenosine is an endogenous signaling molecule upregulated during inflammatory conditions. Acting through the A2b receptor (A2bR), the predominant adenosine receptor in human colonic epithelia, adenosine has been directly implicated in immune and inflammatory responses in the intestine. Little is known about expression and regulation of A2bR during inflammation. Tumor necrosis factor alpha (TNF-alpha) is highly upregulated during chronic and acute inflammatory diseases. This study examined the expression of A2bR during colitis and studied effects of TNF-alpha on A2bR expression, signaling and function. Results demonstrated that A2bR expression increases during active colitis. TNF-alpha pretreatment of intestinal epithelial cells increased A2bR messenger RNA and protein expression. TNF-alpha significantly increased adenosine-induced membrane recruitment of A2bR and cyclic adenosine monophosphate downstream signaling. Further, TNF-alpha potentiated adenosine-induced shortcircuit current and fibronectin secretion. In conclusion, we demonstrated that TNF-alpha is an important regulator of A2bR, and during inflammation, upregulation of TNF-alpha may potentiate adenosine-mediated responses.


Subject(s)
Colitis/metabolism , Intestinal Mucosa/metabolism , Receptor, Adenosine A2B/biosynthesis , Receptor, Adenosine A2B/genetics , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/physiology , Adenosine/metabolism , Adult , Animals , Cell Line , Chlorides/metabolism , Colitis/chemically induced , Cyclic AMP/metabolism , Dextran Sulfate/administration & dosage , Drug Synergism , Fibronectins/metabolism , Humans , Mice , Mice, Inbred C57BL
5.
Curr Gastroenterol Rep ; 3(5): 392-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11560796

ABSTRACT

The epithelium of the intestinal tract is a key barrier between the external environment and the internal body environment. Intestinal epithelial cells are targets for luminal bacteria and viruses and must discriminate between pathogenic and nonpathogenic commensal organisms. Pathogenic bacteria and their secreted products influence epithelial cell function and induce diarrhea by numerous mechanisms that range from an effect on epithelial cell-cell associations to intracellular signal transduction pathways. These effects lead to an inflammatory response and an influx of neutrophils into the epithelium. Infiltrating neutrophils, in turn, signal to epithelial cells, induce a secretory response, and perpetuate the diarrhea. Conversely, commensal bacteria have the ability to suppress inflammatory responses by inhibiting specific intracellular signal transduction pathways. Some of these diverse host pathogenic responses are addressed in this review.


Subject(s)
Bacteria/immunology , Bacterial Toxins/immunology , Cell Communication/physiology , Intercellular Junctions/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Animals , Cell Communication/immunology , Diarrhea/immunology , Diarrhea/physiopathology , Humans , Immunity/immunology , Inflammation/immunology , Intercellular Junctions/immunology , Intestinal Mucosa/physiopathology , Intestinal Mucosa/virology , Neutrophils/immunology , Signal Transduction/immunology , Signal Transduction/physiology , Viruses/immunology
6.
Gastroenterology ; 120(7): 1666-79, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375948

ABSTRACT

BACKGROUND & AIMS: hPepT1 is an intestinal epithelial apical membrane transporter responsible for uptake of di/tripeptides (including bacterial derived proinflammatory n-formyl peptides). hPepT1 expression normally has a strict axial gradient-highest in the proximal small intestine with no expression in the colon. METHODS: Small intestinal-like cells (Caco2-BBE), and colonic-like cells (HT29-Cl.19A), and colonic mucosa from diseased and control patients were used in the present study. RESULTS: hPepT1 expression occurs aberrantly in the colon with chronic ulcerative colitis (6 patients) and Crohn's disease (4 patients), but not in normal colon (4 patients) or colon with microscopic colitis (4 patients). To model expression of hPepT1 by colonic-like cells in inflamed states, we stably transfected HT29-Cl.19A cells with a modified hPepT1 tagged on the N-terminus with green fluorescence protein. Analysis of transfected cells revealed that: GFP-hPepT1 protein, like the natural protein, is targeted to the apical plasma membrane. In addition, the tagged protein retains the capability of di/tripeptide absorption, and the expression of the tagged protein by HT29-Cl.19A cells permits absorption of N-formyl-methionyl-leucyl-phenylalanine (fMLP), as occurs in hPepT1 expressing Caco2-BBE cells. fMLP uptake by colonic cells expressing GFP-hPepT1 specifically enhances major histocompatibility complex class I surface expression. CONCLUSIONS: These data collectively indicate that, in some states of chronic inflammation, hPepT1 may be anomolously expressed in the colon. Further, transport of fMLP by hPepT1 potentially stimulates expression of key accessory immune molecule, MHC-1.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/physiology , Histocompatibility Antigens Class I/analysis , Inflammatory Bowel Diseases/metabolism , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Symporters , Amino Acid Sequence , Biological Transport , Caco-2 Cells , Carrier Proteins/analysis , Colitis/metabolism , Colon/chemistry , HT29 Cells , Humans , Intestine, Small/chemistry , Molecular Sequence Data , Peptide Transporter 1
7.
J Clin Invest ; 107(7): 861-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285305

ABSTRACT

Adenosine is formed in the intestinal lumen during active inflammation from neutrophil-derived 5' AMP. Using intestinal epithelial cell line T84, we studied the effect of adenosine on the secretion of IL-6, a proinflammatory cytokine involved in neutrophil degranulation and lymphocyte differentiation. Stimulation of T84 monolayers with either apical or basolateral adenosine induces A2b receptor-mediated increase in IL-6 secretion, which is polarized to the apical (luminal) compartment. In addition, Salmonella typhimurium, TNF-alpha, and forskolin, known inducers of IL-6 secretion in intestinal epithelial cells, also stimulate IL-6 secretion into the apical compartment. We show that IL6 promoter induction by adenosine occurs through cAMP-mediated activation of nuclear cAMP-responsive element-binding protein (CREB). We also show that IL-6 released in the luminal (apical) compartment achieves a sufficient concentration to activate neutrophils (from which the adenosine signal originates), since such IL-6 is found to induce an intracellular [Ca(++)] flux in neutrophils. We conclude that adenosine released in the intestinal lumen during active inflammation may induce IL-6 secretion, which is mediated by cAMP/CREB activation and occurs in an apically polarized fashion. This would allow sequential activation of neutrophil degranulation in the lumen -- a flow of events that would, in an epithelium-dependent fashion, enhance microbicidal activity of neutrophils as they arrive in the intestinal lumen.


Subject(s)
Adenosine/metabolism , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Neutrophils/metabolism , Signal Transduction/physiology , Activating Transcription Factors , Adenosine/pharmacology , Animals , Blood Proteins/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Colforsin/metabolism , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Intestinal Mucosa/cytology , Purinergic P1 Receptor Antagonists , Receptor, Adenosine A2B , Receptors, Purinergic P1/metabolism , Salmonella typhimurium/metabolism , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
Curr Opin Investig Drugs ; 2(10): 1395-400, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11890354

ABSTRACT

Genetics Institute has developed and launched oprelvekin (rhIL-11; Neumega), a recombinant form of human IL-11. In November 1997, the FDA cleared oprelvekin for the prevention of severe thrombocytopenia and the reduction of the need for platelet transfusions following myelosuppressive chemotherapy in susceptible patients with non-myeloid malignancies 12703021. The product was launched at the end of 1997 [312556]. By December 1999, phase III trials for Crohn's disease (CD) were underway [363007]. Genetics Institute had commenced a 150-patient phase II trial for mild-to-moderate CD and mucositis and the company planned to file regulatory procedures for the indication of CD in 1999 [271210]. An oral formulation for this indication has been developed. Oprelvekin is also undergoing phase I clinical trials for colitis [396157], phase II clinical trials for rheumatoid arthritis [413835] and clinical trials for psoriasis [299644]. In March 1997, Wyeth-Ayerst became the licensee for Europe, Africa, Latin America and Asia (with the exception of Japan). Genetics Institute holds marketing rights for North America [239273]. In Japan, oprelvekin is being developed by Genetics Institute and Yamanouchi; phase III trials have commenced [295049] and were ongoing in May 2001 [411763]. In April 1996, analysts at Yamaichi estimated launch in 2001 and maximum annual sales of over yen 10 billion [215896]. In January 1998, Morgan Stanley Dean Witter predicted Yamanouchi's share of sales to be yen 1 billion in 2001, rising to yen 2 billion in 2002 [315458]. Sales of oprelvekin were US $34 million for Genetics institute in fiscal 2000 while, in July 2001, Credit Suisse First Boston estimated that this figure will be US $30 million and US $34 million in 2001 and 2002, respectively [416883].


Subject(s)
Arthritis, Rheumatoid/drug therapy , Inflammatory Bowel Diseases/drug therapy , Interleukin-11/therapeutic use , Recombinant Proteins/therapeutic use , Thrombocytopenia/drug therapy , Animals , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Interleukin-11/adverse effects , Interleukin-11/metabolism , Interleukin-11/pharmacology , Interleukin-11/toxicity , Recombinant Proteins/adverse effects , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , Structure-Activity Relationship
9.
Am J Physiol Cell Physiol ; 278(6): C1230-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10837351

ABSTRACT

UNLABELLED: It is not known if, in polarized cells, desensitization events can be influenced by the domain on which the receptor resides. Desensitization was induced by 5'-(N-ethylcarboxamido)adenosine (NECA) and was quantitated by measurement of short-circuit current (I(sc)) in response to adenosine. NECA added to either the apical or basolateral compartments rapidly desensitized receptors on these respective domains. Although apical NECA had no effect on the basolateral receptor stimulation, basolateral NECA induced a complete desensitization of the apical receptor. We hypothesized that desensitization of apical receptor by basolateral desensitization could relate to a trafficking step in which A2b receptor is first targeted basolaterally upon synthesis and transported to the apical surface via vesicular transport/microtubules. Because desensitization is associated with downregulation of receptors, apical adenosine receptor can thus be affected by basolateral desensitization. Both low temperature and nocodazole inhibited I(sc) induced by apical and not basolateral adenosine. IN CONCLUSION: 1) a single receptor subtype, here modeled by the A2b receptor, differentially desensitizes based on the membrane domain on which it is expressed, 2) agonist exposure on one domain can result in desensitization of receptors on the opposite domain, 3) cross-domain desensitization can display strict polarity, and 4) receptor trafficking may play a role in the cross-desensitization process.


Subject(s)
Cell Membrane/physiology , Epithelial Cells/cytology , Receptors, Purinergic P1/physiology , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Cell Line , Cell Polarity , Epithelial Cells/physiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nocodazole/pharmacology , Receptor, Adenosine A2B , Receptors, Purinergic P1/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...