Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 5(5): e230, 2016 May 30.
Article in English | MEDLINE | ID: mdl-27239962

ABSTRACT

Thrombospondin 1 is a glycoprotein that regulates cellular phenotype through interactions with its cellular receptors and extracellular matrix-binding partners. Thrombospondin 1 locally regulates angiogenesis and inflammatory responses that contribute to colorectal carcinogenesis in Apc(Min/+) mice. The ability of thrombospondin 1 to regulate responses of cells and tissues to a variety of stresses suggested that loss of thrombospondin 1 may also have broader systemic effects on metabolism to modulate carcinogenesis. Apc(Min/+):Thbs1(-/-) mice exhibited decreased survival and higher tumor multiplicities in the small and large intestine relative to Apc(Min/+) mice when fed a low (5%) fat western diet. However, the protective effect of endogenous thrombospondin 1 was lost when the mice were fed a western diet containing 21% fat. Biochemical profiles of liver tissue identified systemic metabolic changes accompanying the effects of thrombospondin 1 and dietary lipid intake on tumorigenesis. A high-fat western diet differentially regulated elements of amino acid, energy and lipid metabolism in Apc(Min/+):Thbs1(-/-) mice relative to Apc(Min/+):Thbs1(+/+)mice. Metabolic changes in ketone body and tricarboxylic acid cycle intermediates indicate functional interactions between Apc and thrombospondin 1 signaling that control mitochondrial function. The cumulative diet-dependent differential changes observed in Apc(Min/+):Thbs1(-/-) versus Apc(Min/+) mice include altered amino acid and lipid metabolism, mitochondrial dysfunction, eicosanoids and ketone body formation. This metabolic profile suggests that the protective role of thrombospondin 1 to decrease adenoma formation in Apc(Min/+) mice results in part from improved mitochondrial function.

2.
Oncol Rep ; 13(6): 1017-27, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15870916

ABSTRACT

Clinical, epidemiological and experimental findings have provided evidence supporting a role of free radicals in the etiology of cancer. Free radical production is enhanced in many disease states, by carcinogen exposure, and under conditions of stress contributing widely to cancer development in humans. We have established an experimental breast cancer model to examine the effects of all-trans-retinol (retinol/vitamin A) on the production of free radicals in human breast epithelial cells induced by high linear energy transfer (LET)-radiation in the presence of 17beta estradiol. The following cell lines were used in these studies: the MCF-10F cell line, a spontaneously immortalized human breast epithelial cell line. Alpha 5 derived from MCF-10F cells irradiated with two separated doses of 60 cGy alpha particles in the presence of estrogens (60E/60E). Tumor 2, from a tumor formed in nude mice after injection with the cell line alpha 5. Tumor 3, from secondary tumor formed from injecting tumor 2 cells into nude mice. Each of the cell types examined had significantly elevated H(2)O(2) production levels compared to MCF-10F control cells (p<0.001). Retinol (1 microl/ml) significantly (p<0.05) decreased H(2)O(2) production in all cell types examined. Retinol significantly decreased (p<0.05) invasive capabilities of cells across matrigel coated invasion chambers and significantly reduced (p<0.05) PCNA, Fra-1, mutant p53 and increased Rb protein expression levels in comparison to non-retinol-treated ones when assayed using immunofluorescent staining coupled with confocal microscopy. The reduced H(2)O(2) production, decrease in cell invasive capabilities and alterations in protein expression levels suggest that retinol can be used as a chemopreventive agent in human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Estradiol/pharmacology , Vitamin A/pharmacology , Animals , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/radiation effects , Humans , Hydrogen Peroxide/metabolism , Linear Energy Transfer , Mice , Mice, Nude , Mutation , Neoplasm Invasiveness/pathology , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
3.
J Nutr ; 131(11 Suppl): 3041S-6S, 2001 11.
Article in English | MEDLINE | ID: mdl-11694645

ABSTRACT

High intakes of dietary fiber or resistant starches have been associated with a lower incidence of colon cancers. Because short-chain fatty acids (SCFA) such as butyrate are produced in the colonic lumen by the bacterial fermentation of dietary fibers and resistant starches, we hypothesized that SCFA may inhibit the development of invasive human colon cancers. To test this hypothesis, primary human invasive colonocytes were isolated from fresh surgical specimens and treated with 0.01 mol/L acetate, propionate or butyrate; cell invasion, cell adhesion, F-actin polymerization, urokinase plasminogen activator (uPA), tissue inhibitor matrix metalloproteinase (TIMP)-1, TIMP-2 and mutant p53, Bcl-2, Bax, p21 and proliferating cell nuclear antigen (PCNA) protein expression levels were examined. Although each of the SCFA tested significantly reduced primary cell invasion, butyrate was the most potent, inhibiting primary invasive human colon cancer invasion by 54% (P < 0.0001). The effects of SCFA on primary cell invasion appeared to be independent of cell adhesion and F-actin polymerization but dependent on the inhibition of uPA (P < 0.05) and the stimulation of TIMP-1 and TIMP-2 activities (P < 0.05). Protein expression levels of mutant p53, p21, Bax, Bcl-2 and PCNA were significantly altered by each of the SCFA tested (P < 0.05). These data indicate that SCFA inhibit invasive human colon cancer by modulating proteolytic uPA and antiproteolytic TIMP-1 and TIMP-2 activities, but their mechanisms of action on tumor suppression, apoptosis and growth arrest may differ.


Subject(s)
Adenocarcinoma/prevention & control , Cell Adhesion/drug effects , Colonic Neoplasms/prevention & control , Fatty Acids, Volatile/pharmacology , Actins/chemistry , Actins/drug effects , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Cell Division/drug effects , Cell Division/genetics , Cell Movement/drug effects , Colon/cytology , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Dietary Fiber/metabolism , Female , Fermentation , Genes, Tumor Suppressor/drug effects , Humans , Male , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasm Invasiveness , Polymers , Tissue Inhibitor of Metalloproteinases/drug effects , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
4.
Dig Dis Sci ; 46(1): 96-105, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11270800

ABSTRACT

Short chain fatty acids may protect colonic mucosa against neoplastic transformation by modulating colonocyte phenotype, DNA synthesis, and c-myc levels. To test this hypothesis, nonmalignant and malignant human colonocytes were isolated from surgical specimens and treated with 10 mM acetate, propionate, or butyrate. Markers of cellular phenotype, DNA synthesis, and c-myc protein levels were assayed by alkaline phosphatase and dipeptidyl dipeptidase IV activities, [3H]thymidine labeling, and western blotting, respectively. Butyrate, in particular, exerted discordant effects on alkaline phosphatase (P < 0.05), and c-myc levels (P < 0.05, N > or = 6) in nonmalignant and malignant human colonocytes. DPDD was unaffected by any of the short chain fatty acids tested. [3H]Thymidine labeling was differentially stimulated by short chain fatty acids in both cell types and greater DNA synthesis rates were observed in malignant colonocytes (P < 0.005, N = 16). These data suggest that in vitro, butyrate, in particular, may differentially modulate phenotype, DNA synthesis, and c-myc in nonmalignant and malignant human colonocytes.


Subject(s)
Colon/cytology , Colon/pathology , Colonic Neoplasms/pathology , DNA/biosynthesis , Fatty Acids, Volatile/pharmacology , Proto-Oncogene Proteins c-myc/analysis , Aged , Alkaline Phosphatase/analysis , Butyrates/pharmacology , Cell Separation , Cell Survival , Cells, Cultured , DNA, Neoplasm/biosynthesis , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/analysis , Female , Humans , Male , Phenotype , Stimulation, Chemical , Thymidine/metabolism
5.
J Gastrointest Surg ; 4(5): 501-12, 2000.
Article in English | MEDLINE | ID: mdl-11077326

ABSTRACT

Short-chain fatty acids (SCFAs) butyrate, propionate, and acetate produced during fiber fermentation promote colonic differentiation and can reverse or suppress neoplastic progression. We sought to identify candidate genes responsible for SCFA activity on colonocytes and to compare the relative activities of independent SCFAs. cDNA was generated from polyA+ mRNA isolated from control Caco-2 cells and cells treated with equimolar butyrate, propionate, and acetate. GeneCalling, a restriction-based differential RNA expression platform linked to a DNA sequence database lookup, was applied. A total of 30,000 individual genetic sequences were analyzed for differential expression among the three SCFAs. Differentially expressed peaks corresponding to cancer-related genes were isolated, sequenced, and cross-referenced to the GenBank human database. Gene identities were independently confirmed by oligonucleotide poisoning. More than 1000 gene fragments were identified as being substantially modulated in expression by butyrate. Butyrate tended to have the most pronounced effects and acetate the least. Five fragments selected for further study were fully sequenced and proved 100% homologous with human sequences for clusterin, amyloid precursor-like protein 2, and caudal homeobox 2 protein, not previously known to be modulated by SCFAs. In each case, a similar order of potency for the three SCFAs studied was observed. The common SCFAs appear to exert different effects. This study suggests the diversity of the SCFA response at the molecular level and facilitates identifying genes important in the biologic activity of dietary fiber.


Subject(s)
Colon/cytology , Enterocytes/metabolism , Fatty Acids, Volatile/genetics , Gene Expression , Molecular Chaperones , Alzheimer Disease , Amyloid beta-Protein Precursor , Butyrates , Caco-2 Cells , Clusterin , Dietary Fiber , Genes, Homeobox , Glycoproteins/genetics , Humans , Nerve Tissue Proteins
6.
Cell Physiol Biochem ; 10(1-2): 27-36, 2000.
Article in English | MEDLINE | ID: mdl-10844395

ABSTRACT

BACKGROUND/AIMS: Cell-matrix interactions influence intestinal epithelial biology, but the whether specific integrin heterodimers exert different effects is unclear. METHODS: We used functional antibodies to investigate effects of the alpha2, alpha3, alpha5, and alpha6 integrin subunits on proliferation and differentiation of human intestinal Caco-2 cells on laminin. Cells seeded onto laminin-coated inserts in defibronectinized medium were treated with functional antibodies or normal IgG for 72 hrs and proliferation, alkaline phosphatase and dipeptidyl dipeptidase specific activity were measured. RESULTS: Caco-2 adhesion to antibody to each alpha-integrin subunit stimulated tyrosine phosphorylation of Focal Adhesion Kinase and paxillin, suggesting that these antibodies intiate integrin-related tyrosine signalling. Proliferation was inhibited by anti-alpha2 and anti-alpha3, but stimulated by anti-alpha6. Alkaline phosphatase and dipeptidyl dipeptidase specific activity were promoted by alpha2 blockade but decreased after a6 blockade. Proliferative blockade using mitomycin C or hydroxyurea prevented the effects of alpha2 ligation on differentiation, but the decrease in alkaline phosphatase specific activity observed after a6 integrin subunit blockade was preserved even after proliferative blockade. CONCLUSION: lntegrin heterodimers modulate human Caco-2 intestinal epithelial biology on laminin in an alpha-subunit specific manner. The different effects of anti-alpha2 and anti-alpha6 may reflect competitition by these antibodies with integrin subunit interactions with laminin as well as initiation of their own signals or different functions for the alpha6 integrin subunit


Subject(s)
Cell Division/physiology , Integrins/physiology , Intestines/cytology , Antibodies/immunology , Caco-2 Cells , Epithelial Cells/cytology , Humans , Integrins/immunology , Phenotype , Signal Transduction/physiology
7.
J Surg Res ; 76(1): 41-6, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9695737

ABSTRACT

BACKGROUND: Short chain fatty acids derived from dietary fiber may protect against invasive colon cancer by modulating degradative matrix metalloproteinases (MMPs) and protective tissue inhibitor matrix metalloproteinases (TIMPs). Since invasion depends on the MMP/TIMP ratio, we hypothesized that short chain fatty acids inhibit colon cancer invasion by inhibiting MMPs and stimulating TIMPs. MATERIALS AND METHODS: SW1116 colon cancer cells were seeded onto Matrigel-coated Boyden chambers and treated with unsupplemented media or media containing 10 mM acetate, propionate, or butyrate. SW1116 invasion was quantitated by light microscopy and conditioned media were assayed by ELISA for MMP-1,2,3,9; TIMP-1,2; MMP/TIMP complex; and urokinase plasminogen activator (uPA). All data are expressed as mean percentage of control +/- SE (n > 6). RESULTS: Although all three short chain fatty acids inhibited invasion, butyrate was more potent than either acetate or propionate, inhibiting SW1116 invasion by 35 +/- 1% of control (n = 18, P < .0001) vs. 18 +/- 9% (n = 7, P < .05) for acetate and 10 +/- 6% (n = 7, P < .05) for propionate. MMP-2 was not modulated by any of the short chain fatty acids while MMP-1 was modulated only by butyrate and MMP-3 by propionate. Acetate did not modulate MMPs, TIMP-1, or uPA, but stimulated TIMP-2. In contrast, propionate and butyrate stimulated MMP-9 and TIMP-2 by 119-233% and both inhibited uPA by 8-16%. TIMP-1 was stimulated only by butyrate and actually inhibited by propionate. Only butyrate stimulated both TIMP-1 and TIMP-2. CONCLUSIONS: These data suggest that dietary fiber may protect against invasive colon cancer through stimulation of TIMP and inhibition of uPA activities, rather than through short chain fatty acids effects on the activities of the MMPs studied.


Subject(s)
Fatty Acids, Volatile/pharmacology , Neoplasm Invasiveness/physiopathology , Protease Inhibitors/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Acetates/pharmacology , Adenocarcinoma , Butyrates/pharmacology , Cell Division/drug effects , Colonic Neoplasms , Dietary Fiber/metabolism , Enzyme Activation/drug effects , Humans , Metalloendopeptidases/metabolism , Propionates/pharmacology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
8.
Cell Tissue Res ; 292(3): 553-62, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9582412

ABSTRACT

Intestinal epithelial cell differentiation is closely regulated during normal cell renewal, maturation, and malignant transformation. Since tyrosine phosphorylation influences differentiation in other cell types and has been reported to vary between crypt cells to differentiated villus tip cells, we investigated the influence of tyrosine phosphorylation in colonocyte differentiation, by using human colonic Caco-2 cells as a model and expression of the brush border enzymes alkaline phosphatase (AKP) and dipeptidyl peptidase (DPDD) as differentiation markers. We studied three tyrosine kinase inhibitors with different modes of action and specificities, viz., genistein, erbstatin analog (EA), and tyrphostin, and the tyrosine phosphatase inhibitor sodium orthovanadate. AKP- and DPDD-specific activities were assayed in protein-matched cell lysates by synthetic substrate digestion. We also correlated the effects of these agents on brush border enzyme activity with tyrosine phosphorylation of phosphoproteins by Western blotting. Genistein (5-75 mg/ml) dose-dependently stimulated AKP and DPDD with a maximal stimulation at 75 mg/ml by 158.6+/- 17.5% and 228.6+/-37.1% of control values, respectively (n=12, P<0.001). The inactive analog genistin had no effect. Tyrphostin (25 mM) similarly stimulated AKP and DPDD by 138. 6+/-6.6% and 131.8+/-1.5% of control values (n=12, P<0.001). Unexpectedly, EA (0.1-10 mM) had the opposite effect, inhibiting AKP- and DPDD-specific activity significantly at 10 mM with a maximal 14.8+/-6.4% and 26.5+/-2.5% of control values (n=12, each P<0.001). Sodium orthovanadate had a discordant effect on these two differentiation markers. Orthovanadate dose-dependently increased AKP to a maximal 188.5+/-16.1% of basal activity at 1.5 mM but decreased DPDD activity at 1.5 mM to 47.2+/-3.8% (n=9, P<0.001 each). The effects of each agent were preserved when proliferation was blocked with mitomycin C, suggesting that the modulation of phenotype by these agents was independent of any effects of proliferation. The tyrosine phosphorylation of several phosphoprotein bands was affected differently by these agents. In particular, the tyrosine phosphorylation of one 70-kDa to 71-kDa band was increased by genistein and tyrophostin but deceased by EA. The different effects of these modulators of tyrosine kinase activity raise the possibility that at least two independent enzymes or pathways regulating tyrosine phosphorylation modulate intestinal epithelial differentiation. Furthermore, tyrosine phosphorylation of the 70-kDa to 71-kDa phosphoprotein may be important in the intracellular signaling by which intestinal epithelial cell differentiation is controlled.


Subject(s)
Caco-2 Cells/cytology , Caco-2 Cells/enzymology , Tyrosine/metabolism , Tyrphostins , Alkaline Phosphatase/metabolism , Caco-2 Cells/drug effects , Catechols/pharmacology , Cell Differentiation/physiology , Cell Division/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Humans , Hydroquinones/pharmacology , Microvilli/drug effects , Microvilli/enzymology , Microvilli/ultrastructure , Nitriles/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Second Messenger Systems/physiology , Vanadates/pharmacology
9.
Proc Soc Exp Biol Med ; 217(4): 476-83, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9521097

ABSTRACT

Fermentation of dietary fiber within the colonic lumen yields short chain fatty acids (SCFA) such as butyrate, which may modulate colonic mucosal biology and inhibit the development of a malignant phenotype. However, different fibers yield varying proportions of various SCFA. We studied the effects of the three most common SCFA, acetate, butyrate, and propionate, on the proliferation, adhesion, and motility of the human intestinal Caco-2 cell line, as well as the effects of these SCFA on alkaline phosphatase and dipeptidyl dipeptidase specific activity (common laboratory markers of differentiation). In addition, we examined the modulation of c-myc protein and the tyrosine phosphorylation of cellular proteins by these SCFA in order to determine whether the variations in the potency of these three SCFA for phenotypic change extended to variations in effects on intracellular signaling and protooncogene expression. All three SCFA tended to slow proliferation, promote brush border enzyme activity, and inhibit both adhesion to and motility across a type I collagen matrix substrate. However, we observed substantial differences in the potency of these three SCFA with regard to these effects. In particular, butyrate was uniformly more potent than an equimolar concentration of acetate whereas equimolar propionate achieved comparable effects with regard to proliferation and brush border enzyme activity but was intermediate between butyrate and acetate with regard to modulation of cell-matrix interactions. Similarly, the SCFA downregulated c-myc protein levels and modulated the phosphorylation of several intracellular tyrosine phosphoproteins, but the effects of the three SCFA varied substantially for these parameters. These results suggest that the common short chain fatty acids are not equipotent in their effects on human Caco-2 colon cancer cell biology. Such differences in potency could contribute to the observed differences in effects of different dietary fibers in vivo.


Subject(s)
Caco-2 Cells/drug effects , Fatty Acids, Volatile/pharmacology , Intestinal Mucosa/drug effects , Acetates/pharmacology , Alkaline Phosphatase/metabolism , Blotting, Western , Butyrates/pharmacology , Butyric Acid , Caco-2 Cells/cytology , Cell Division/drug effects , Cell Movement/drug effects , Humans , Intestinal Mucosa/cytology , Phenotype , Phosphorylation , Phosphotyrosine/metabolism , Propionates/pharmacology , Proto-Oncogene Proteins c-myc/metabolism
10.
Digestion ; 58(1): 34-42, 1997.
Article in English | MEDLINE | ID: mdl-9018008

ABSTRACT

Sucralfate, which binds to the matrix of the ulcer bed, is theoretically advantageous for duodenal ulcer therapy, but has not fulfilled its promise clinically. We examined the effects of sucralfate and related compounds in a human intestinal epithelial (Caco-2) cell culture model of restitution on sheet migration across and adhesion to collagen I. Migration was quantitated across a collagen I matrix treated with sucralfate or related compounds and correlated with cell adhesion. Caco-2 motility was significantly and dose-responsively inhibited by sucralfate at therapeutic luminal concentrations. Sucrose octaacetate, the sucralfate backbone, and lactose octaacetate exhibited similar effects while the beta-bonded disaccharide maltose octaacetate had little effect. Sucrose itself slightly stimulated motility. Adhesion effects paralleled motility. Thus, sucralfate may inhibit intestinal epithelial motility by sterically interfering with adhesion to collagen I. A sucralfate analog with a lactose octaacetate backbone might retain growth factor binding without inhibiting enterocyte motility, perhaps improving its clinical efficacy.


Subject(s)
Anti-Ulcer Agents/pharmacology , Caco-2 Cells/cytology , Cell Movement/drug effects , Intestinal Mucosa/cytology , Sucralfate/pharmacology , Antibiotics, Antineoplastic/pharmacology , Caco-2 Cells/drug effects , Cell Adhesion/drug effects , Collagen/metabolism , Dose-Response Relationship, Drug , Humans , Intestinal Mucosa/drug effects , Mitomycin/pharmacology , Sucrose/analogs & derivatives , Sucrose/pharmacology , Wound Healing/drug effects , Wound Healing/physiology
11.
Am J Clin Nutr ; 64(5): 757-60, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8901797

ABSTRACT

Previous work has suggested that kidney hemodialysis patients could be at risk for either moderate copper deficiency or copper toxicity. The present study examined copper-related blood indexes in subjects undergoing hemodialysis treatments with membranes that are not copper-based, in subjects undergoing chronic ambulatory peritoneal dialysis (CAPD), and in control subjects. Both dialysis groups had low plasma copper and ceruloplasmin activities. This occurred despite high plasma interleukin 6 concentrations, a situation that usually elevates plasma ceruloplasmin and copper values. CAPD and hemodialysis subjects had low ratios of ceruloplasmin activity to immunoreactive protein, and low ratios of plasma copper to ceruloplasmin protein. Both are signs of copper deficiency. In contrast, copper-containing erythrocyte superoxide dismutase (SOD) activities were high in hemodialysis subjects and showed a nonsignificant trend toward high values in CAPD subjects. Blood mononuclear cell copper contents were highly variable within each group, and there were no significant differences between groups. In conclusion, ceruloplasmin-related indexes in kidney dialysis patients not dialyzed with copper-based membranes suggested a tendency toward moderate copper deficiency. However, this contention could not be confirmed by erythrocyte SOD activity or mononuclear cell copper measurements.


Subject(s)
Copper/blood , Kidney Diseases/blood , Kidney Diseases/therapy , Peritoneal Dialysis, Continuous Ambulatory , Adult , Aged , Ceruloplasmin/analysis , Copper/analysis , Erythrocytes/enzymology , Female , Humans , Interleukin-6/blood , Kidney/physiology , Male , Middle Aged , Monocytes/chemistry , Superoxide Dismutase/analysis , Superoxide Dismutase/blood
12.
Exp Cell Res ; 225(2): 301-5, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8660918

ABSTRACT

Extracellular matrix regulation of intestinal epithelial differentiation may affect development, differentiation during migration to villus tips, healing, inflammatory bowel disease, and malignant transformation. Cell culture studies of intestinal epithelial biology may also depend on the matrix substrate used. We evaluated matrix effects on differentiation and proliferation in human intestinal Caco-2 epithelial cells, a model for intestinal epithelial differentiation. Proliferation, brush border enzyme specific activity, and spreading were compared in cells cultured on tissue culture plastic with interstitial collagen I and the basement membrane constituents collagen IV and laminin. Each matrix significantly increased alkaline phosphatase, dipeptidyl peptidase, lactase, sucrase-isomaltase, and cell spreading in comparison to plastic. However, the basement membrane proteins collagen IV and laminin further promoted all four brush border enzymes but inhibited spreading compared to collagen I. Proliferation was most rapid on type I collagen and slowest on laminin and tissue culture plastic. Basement membrane matrix proteins may promote intestinal epithelial differentiation and inhibit proliferation compared with interstitial collagen I.


Subject(s)
Caco-2 Cells/cytology , Extracellular Matrix Proteins/pharmacology , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Caco-2 Cells/drug effects , Caco-2 Cells/enzymology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Size/drug effects , Collagen/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/drug effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Laminin/pharmacology , Oligo-1,6-Glucosidase/drug effects , Oligo-1,6-Glucosidase/metabolism , beta-Galactosidase/drug effects , beta-Galactosidase/metabolism
13.
Yale J Biol Med ; 69(2): 119-29, 1996.
Article in English | MEDLINE | ID: mdl-9112743

ABSTRACT

Intestinal epithelial cells migrating across a mucosal defect are generally described as dedifferentiated, a term that suggests a loss of regulatory biology. Since cell biology may be more readily studied in established cell lines than in vivo, a model is developed using the human Caco-2 intestinal epithelial cell migrating across matrix proteins. This resembles in vivo models of mucosal healing in its sheet migration and loss of the brush border enzymes, which are conventional markers for intestinal epithelial differentiation. Immunohistochemical studies of migrating Caco-2 cells suggest, however, that the rearrangements of cytoskeletal, cell-cell and cell-matrix proteins during migration are not random but seem adapted to the migratory state. Indeed, Caco-2 migration may be substantially regulated by a variety of physiologic and pharmacologic stimuli and differentiation, measured by the specific activity of the intestinal epithelial brush border enzymes alkaline phosphatase and dipeptidyl dipeptidase, may be independently pharmacologically programmed during the stimulation or inhibition of cell motility.


Subject(s)
Cell Movement/physiology , Intestinal Mucosa/pathology , Intestines/cytology , Microvilli/enzymology , Actins/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Collagen/chemistry , Collagen/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/drug effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Enterostomy , Epidermal Growth Factor/pharmacology , Epithelial Cells , Epithelium/pathology , Epithelium/ultrastructure , Humans , Hydrogen-Ion Concentration , Intestinal Mucosa/cytology , Intestines/ultrastructure , Jejunum/pathology , Jejunum/surgery , Laminin/metabolism , Membrane Proteins/metabolism , Microvilli/drug effects , Pentagastrin/pharmacology , Peptide YY , Peptides/pharmacology , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Zonula Occludens-1 Protein
14.
J Surg Res ; 59(1): 121-6, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7630114

ABSTRACT

Much is known about intestinal epithelial regulation by growth factors and nutrients but the intracellular signals governing cell phenotype are less well understood. In an initial attempt to evaluate the role of protein kinase C in these events, we studied the effects of protein kinase C modulation by the phorbol ester TPA upon the differentiation, motility, and doubling time of the human intestinal epithelial Caco-2 cell line, a common model for enterocytic brush border enzyme expression. We also compared the effects of TPA to those of 4 alpha-phorbol 12,13-didecanoate, which does not modulate protein kinase C activity. Differentiation was studied by quantitating brush order dipeptidyl peptidase (DPDD)-specific activity in protein-matched Caco-2 lysates via synthetic substrate digestion. Alkaline phosphatase (AP) was studied for comparison. Doubling time was assessed by log transformation of serial cell counts and motility by monolayer expansion across type I collagen. TPA (0.03-0.7 micrograms/ml) dose-dependently stimulated DPDD, with a maximal 455 +/- 26% increase at 0.7 micrograms/ml (P < 0.01, n = 5). However, TPA dose-dependently inhibited AP to a maximal 91.6 +/- 0.3% decrease (P < 0.01, n = 5). TPA also dose-dependently prolonged the cell doubling time from 26.5 +/- 0.4 to 64.5 +/- 8.8 hr (n = 20, P < 0.01) with a maximal effect at 1.0 micrograms/ml and inhibited migration with essentially complete ablation of cell motility at 0.1 micrograms/ml (n = 10, P < 0.001).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Intestines/enzymology , Protein Kinase C/physiology , Tetradecanoylphorbol Acetate/pharmacology , Alkaline Phosphatase/metabolism , Cell Line , Cell Movement , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Dose-Response Relationship, Drug , Humans , Intestines/cytology , Microvilli/enzymology , Phorbol Esters/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...