Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Ann Oncol ; 29(11): 2240-2246, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30247524

ABSTRACT

Background: The consensus molecular subtypes (CMS) is a transcriptome-based classification of colorectal cancer (CRC) initially described in early-stage cohorts, but the associations of CMS with treatment outcomes in the metastatic setting are yet to be established. This study aimed to evaluate the prognostic impact of CMS classification and its predictive effects for bevacizumab benefit in metastatic CRC by correlative analysis of the AGITG MAX trial. Patients and methods: The MAX trial previously reported improved progression-free survival (PFS) for the addition of bevacizumab (B) to chemotherapy [capecitabine (C)±mitomycin (M)]. Archival primary tumours from 237 patients (50% of trial population) underwent gene expression profiling and classification into CMS groups. CMS groups were correlated to PFS and overall survival (OS). The interaction of CMS with treatment was assessed by proportional hazards model. Results: The distribution of CMS in MAX were CMS1 18%, CMS2 47%, CMS3 12%, CMS4 23%. CMS1 was the predominant subtype in right-sided primary tumours, while CMS2 was the predominant subtype in left-sided. CMS was prognostic of OS (P = 0.008), with CMS2 associated with the best outcome and CMS1 the worst. CMS remained an independent prognostic factor in a multivariate analysis. There was a significant interaction between CMS and treatment (P-interaction = 0.03), for PFS, with hazard ratios (95% CI) for CB+CBM versus C arms in CMS1, 2, 3 and 4: 0.83 (0.43-1.62), 0.50 (0.33-0.76), 0.31 (0.13-0.75) and 1.24 (0.68-2.25), respectively. Conclusions: This exploratory study found that CMS stratified OS outcomes in metastatic CRC regardless of first-line treatment, with prognostic effects of CMS groups distinct from those previously reported in early-stage cohorts. In CMS associations with treatment, CMS2 and possibly CMS3 tumours may preferentially benefit from the addition of bevacizumab to first-line capecitabine-based chemotherapy, compared with other CMS groups. Validation of these findings in additional cohorts is warranted. Clinical trial number: This is a molecular sub-study of MAX clinical trial (NCT00294359).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Colorectal Neoplasms/drug therapy , Transcriptome/genetics , Adult , Aged , Aged, 80 and over , Capecitabine/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mitomycin/therapeutic use , Prognosis , Progression-Free Survival
2.
Br J Cancer ; 112(2): 313-8, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25412235

ABSTRACT

BACKGROUND: Telomeres are TTAGGG tandem repeats capping chromosomal ends and partially controlled by the telomerase enzyme. The EGFR pathway putatively regulates telomerase function, prompting an investigation of telomere length (TL) and its association with anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer (mCRC). METHODS: Colorectal cancer cell lines were treated with multiple drugs and sensitivity determined. Clinical information was gathered from 75 patients who had received anti-EGFR drugs. Telomere length was measured using a validated qRT-PCR technique. RESULTS: In CRC cell lines, TL independently predicted cetuximab sensitivity. Cells with shorter TL had growth inhibition of 18.6±3.41% as compared with 41.39±8.58% in longer TL (P=0.02). These in vitro findings were validated clinically, in a robust multivariate model. Among patients with KRas WT tumours, those with longer TL had a superior median progression-free survival (PFS) of 24.9 weeks than those with shorter TL; median 11.1 weeks, HR 0.31; P=0.048. CONCLUSION: Telomere length could be a potential unique biomarker predictive of clinical benefit (PFS) of mCRC patients treated with anti-EGFR therapy. This is the novel demonstration of a complex hitherto undescribed interaction, placing anti-EGFR therapy, EGFR pathway, and the telomerase complex within a clinical context.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Liver Neoplasms/drug therapy , Telomere Shortening , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cetuximab , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Middle Aged , Molecular Targeted Therapy , Panitumumab , Telomere
3.
Br J Cancer ; 113(1): 37-45, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26125443

ABSTRACT

BACKGROUND: Bevacizumab prolongs progression-free survival (PFS) in patients with metastatic colorectal cancer. We analysed the protein expression levels of vascular endothelial growth factor (VEGF) ligands and receptors to determine their prognostic and predictive effects. METHODS: We graded expression of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-R1, and VEGF-R2 to assess whether overexpression predicted bevacizumab resistance in samples from 268 of 471 patients randomised to capecitabine (C), capecitabine and bevacizumab (CB), or CB and mitomycin (CBM) in the MAX trial and extended the analysis to the CAIRO-2 population. RESULTS: Patients with low expression of VEGF-D (0, 1þ) benefited from bevacizumab treatment (PFS hazard ratio (HR) (C vs CBþCBM), 0.21; 95% CI, 0.08­0.55; overall survival (OS) HR, 0.35; 95% CI, 0.13­0.90). Patients with higher VEGF-D expression received less benefit (VEGF-D 2þ PFS HR, 0.67; 95% CI, 0.45­1.00; OS HR, 0.82; 95% CI, 0.52­1.30; VEGF-D 3þ PFS HR, 0.77; 95% CI, 0.50­1.17; OS HR, 1.28; 95% CI, 0.79­2.09) (P interaction o0.05). In CAIRO-2, there was no difference in PFS or OS according to VEGF-D expression. CONCLUSIONS: The predictive value of VEGF-D expression for bevacizumab may depend on the chemotherapy backbone used. Further evaluation is required before clinical utilisation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Vascular Endothelial Growth Factor D/metabolism , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Capecitabine , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Mitomycin/administration & dosage , Neoplasm Metastasis
4.
Br J Cancer ; 107(9): 1514-7, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23033009

ABSTRACT

BACKGROUND: Levels of the pro-tumorigenic prostaglandin PGE(2) are increased in colorectal cancer, previously attributed to increased synthesis through COX-2 upregulation and, more recently, to decreased catabolism. The functionally linked genes 15-prostaglandin dehydrogenase (15-PGDH) and the prostaglandin transporter PGT co-operate in prostaglandin degradation and are downregulated in colorectal cancer. We previously reported repression of 15-PGDH expression by the Wnt/ß-catenin pathway, commonly deregulated during early colorectal neoplasia. Here we asked whether ß-catenin also regulates PGT expression. METHODS: The effect of ß-catenin deletion in vivo was addressed by PGT immunostaining of ß-catenin(-/lox)-villin-cre-ERT2 mouse tissue. The effect of siRNA-mediated ß-catenin knockdown and dnTCF4 induction in vitro was addressed by semi-quantitative and quantitative real-time RT-PCR and immunoblotting. RESULTS: This study shows for the first time that deletion of ß-catenin in murine intestinal epithelium in vivo upregulates PGT protein, especially in the crypt epithelium. Furthermore, ß-catenin knockdown in vitro increases PGT expression in both colorectal adenoma- and carcinoma-derived cell lines, as does dnTCF4 induction in LS174T cells. CONCLUSIONS: These data suggest that ß-catenin employs a two-pronged approach to inhibiting prostaglandin turnover during colorectal neoplasia by repressing PGT expression in addition to 15-PGDH. Furthermore, our data highlight a potential mechanism that may contribute to the non-selective NSAID aspirin's chemopreventive efficacy.


Subject(s)
Aspirin/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Intestinal Mucosa/metabolism , Organic Anion Transporters/biosynthesis , beta Catenin/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Cell Line, Tumor , HCT116 Cells , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Mice , Organic Anion Transporters/genetics , Signal Transduction , beta Catenin/genetics
5.
Cancer Res ; 60(16): 4561-72, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969808

ABSTRACT

The short-chain fatty acid butyrate, produced by microbial fermentation of dietary fiber in the large intestine, is a physiological regulator of major pathways of colonic epithelial cell maturation: cell cycle arrest, lineage-specific differentiation, and apoptosis. Microarray analysis of 8,063 sequences demonstrated a complex cascade of reprogramming of SW620 colonic epithelial cells upon treatment with butyrate characterized by the progressive recruitment of gene sets as a function of time. Comparison with the effects of trichostatin A, in conjunction with differences in the kinetics of alteration of histone acetylation induced by butyrate and trichostatin A, identified subsets of induced and repressed genes likely coordinately regulated by altered histone acetylation. The butyrate response was also compared in detail with that of sulindac, a nonsteroidal anti-inflammatory drug with significant chemopreventive activity for colon cancer, and curcumin, a component of mustard and curry structurally and functionally related to sulindac that also has chemopreventive activity. Although gene clusters were identified that showed similar responses to butyrate and sulindac, the data were characterized by the extensive differences in the effects of the two agents. This was striking for functional classes of genes involved in signaling pathways and in cell cycle progression, although butyrate and sulindac induce a similar G0-G1 arrest, elevation of beta-catenin-Tcf signaling, and apoptotic cascade. As regards cell cycle arrest, the underlying mechanism in response to butyrate was most similar to that of the Caco-2 cell line that had spontaneously undergone a G0-G1 arrest and least similar to the G2-M arrest stimulated by curcumin. Thus, high-throughput microarray analysis of gene expression profiles can be used to characterize and distinguish the mechanisms of response of colonic epithelial cells to physiological and pharmacological inducers of cell maturation. This has important implications for characterization of chemopreventive agents and recognition of potential toxicity and synergies. The data bases, gene clusters, and analyses are available at http:// sequence.aecom.yu.edu/genome/.


Subject(s)
Anticarcinogenic Agents/pharmacology , Butyrates/pharmacology , Colon/physiology , Colonic Neoplasms/prevention & control , Enzyme Inhibitors/pharmacology , Acetylation , Caco-2 Cells/cytology , Caco-2 Cells/drug effects , Caco-2 Cells/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Differentiation/physiology , Colon/cytology , Colon/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Curcumin/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Gene Expression Profiling , Histone Deacetylase Inhibitors , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Kinetics , Multigene Family , Signal Transduction/drug effects , Sulindac/pharmacology
6.
Cancer Res ; 60(23): 6704-13, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11118056

ABSTRACT

We have identified an alternative apoptotic cascade induced in SW620 human colonic carcinoma cells by the protein kinase antagonist staurosporine (stsp). Consistent with its effect in other colonic epithelial cells, stsp induced G2-M arrest and apoptosis of SW620 cells. However, despite the paradigm that growth arrest triggers apoptotic cascades, apoptosis was detected before G2-M arrest. Reports have linked dissipation of the mitochondrial membrane potential (deltapsim) to the initiation of apoptosis and have linked elevation of the deltapsim to the escape from apoptosis However, neither apoptosis nor cell cycle arrest were altered by the collapse of the deltapsim, and increased deltapsim enhanced the initiation of apoptosis but blocked G2-M arrest. Although reactive oxygen species (ROS) have been implicated in some colonic epithelial cell and stsp-induced cascades, neither antioxidants nor the inhibition of RNA or protein synthesis altered apoptosis of SW620 cells. Finally, cytosolic cytochrome c has been linked to activation of caspase-3 and dissipation of the deltapsim. However, caspase-3 activation preceded the accumulation of cytochrome c in the cytosol and was accompanied by transient elevations in both the deltapsim and mitochondria-associated cytochrome c. Therefore, we have identified a distinct apoptotic cascade in SW620 cells that was induced independently of growth arrest, dissipation of the deltapsim, ROS production, or synthesis of de novo RNA or protein, and we have linked its efficient initiation to early elevation of the deltapsim.


Subject(s)
Apoptosis/physiology , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , G2 Phase/physiology , Mitochondria/physiology , Mitosis/physiology , Staurosporine/pharmacology , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Division/physiology , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , Colonic Neoplasms/physiopathology , Cytochrome c Group/metabolism , Cytosol/drug effects , Cytosol/metabolism , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , G2 Phase/drug effects , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Ionophores/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitosis/drug effects , Neoplasm Proteins/biosynthesis , Nigericin/pharmacology , RNA/antagonists & inhibitors , Tumor Cells, Cultured
7.
Cancer Res ; 61(8): 3465-71, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309309

ABSTRACT

The beta-catenin TCF pathway is implicated in the regulation of colonic epithelial cell proliferation, but its role in the regulation of cell differentiation is unknown. The colon carcinoma cell line, Caco-2, spontaneously undergoes G(0)/G(1) cell cycle arrest and differentiates along the absorptive cell lineage over 21 days in culture. In parallel, we show that beta-catenin-TCF activity and complex formation are significantly down-regulated. The down-regulation of beta-catenin-TCF signaling was independent of APC, which we characterized as having a nonsense mutation in codon 1367 in Caco-2 cells, but was associated with a decrease in TCF-4 protein levels. Total beta-catenin levels increased during Caco-2 cell differentiation, although this was attributable to an increase in the membrane, E-cadherin-associated, fraction of beta-catenin. Importantly, down-regulation of beta-catenin-TCF signaling in undifferentiated Caco-2 cells by three different mechanisms, ectopic expression of E-cadherin, wild-type APC, or dominant negative TCF-4, resulted in an increase in the promoter activities of two genes that are well-established markers of cell differentiation, alkaline phosphatase and intestinal fatty acid binding protein. These studies demonstrate, therefore, that in addition to its established role in the regulation of cell proliferation, down-regulation of the beta-catenin-TCF pathway is associated with the promotion of a more-differentiated phenotype in colonic epithelial cells.


Subject(s)
Colon/cytology , Cytoskeletal Proteins/physiology , Signal Transduction/physiology , Trans-Activators , Transcription Factors/physiology , Adenomatous Polyposis Coli Protein , Caco-2 Cells , Cadherins/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Colon/metabolism , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intestinal Absorption/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Promoter Regions, Genetic , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , beta Catenin
8.
Oncogene ; 34(10): 1333-9, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-24632621

ABSTRACT

The proto-oncogene c-Jun is a component of activator protein-1 (AP-1) transcription factor complexes that regulates processes essential for embryonic development, tissue homeostasis and malignant transformation. Induction of gene expression by c-Jun involves stimulation of its transactivation ability and upregulation of DNA binding capacity. While it is well established that the former requires JNK-mediated phosphorylation of S63/S73, the mechanism(s) through which binding of c-Jun to its endogenous target genes is regulated remains poorly characterized. Here we show that interaction of c-Jun with chromatin is positively regulated by protein phosphatase 2A (PP2A) complexes targeted to c-Jun by the PR55α regulatory subunit. PR55α-PP2A specifically dephosphorylates T239 of c-Jun, promoting its binding to genes regulating tumour cell migration and invasion. PR55α-PP2A also enhanced transcription of these genes, without affecting phosphorylation of c-Jun on S63. These findings suggest a critical role for interplay between JNK and PP2A pathways determining the functional activity of c-Jun/AP-1 in tumour cells.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Protein Phosphatase 2/metabolism , Transcription Factor AP-1/metabolism , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Phosphorylation , Protein Binding , Protein Phosphatase 2/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism
9.
Inflamm Bowel Dis ; 2(4): 279-302, 1996.
Article in English | MEDLINE | ID: mdl-23282597

ABSTRACT

: The intestinal epithelium must selectively absorb nutrients but exclude luminal pathogens and pro-inflammatory molecules from host tissues. It is topographically organised with proliferating cells buried deep in the crypts and functionally mature cells lining the surface or villus. This ensures efficient barrier function at the interface and relative protection of stem cells from luminal insults. The rapid turnover of cells necessitates high energy requirements. Luminal glutamine and butyrate are necessary to maintain optimal energy status of the small and large intestinal epithelium, respectively. The passage of macromolecules across the epithelium is selectively impeded by enterocytes and intercellular tight junctions. Multiple factors have been recognised that alter paracellular permeability, but relatively little is known regarding control of transcellular passage of macromolecules. The intestinal epithelium is, however, more than just a physical barrier. It can detoxify xenobiotics. It secretes "protective" factors including immunoglobulins, mucus, trefoil peptides, and defensins into the lumen, while epithelial release of chemokines, cytokines, and other inflammatory mediators may initiate mucosal immunity or inflammation. The epithelial response to injurious stimuli is complex and may lead to events that, for example, enhance barrier function, reduce their susceptibility to injury, or recruit secondary protective mechanisms. The intestinal epithelium rapidly migrates across breeches in its continuity, a process that, in more severe injury, sets up a microenvironment more amenable to efficient regeneration. Repair events are also regulated at multiple levels. Thus, the complexity of the active and passive roles of the epithelial barrier in protecting the organism from its environment is now unfolding. The knowledge gained offers insight into pathogenesis of diseases such as inflammatory bowel disease and may form a rational basis for the future design of novel therapeutic strategies in their management.

10.
Oncogene ; 31(14): 1817-24, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-21874050

ABSTRACT

Fos-related antigen-1 (Fra-1) is a member of the Activator Protein-1 (AP-1) transcription factor superfamily that is overexpressed in a variety of cancers, including colon, breast, lung, bladder and brain. High Fra-1 levels are associated with enhanced cell proliferation, survival, migration and invasion. Despite its frequent overexpression, the molecular mechanisms that regulate the accumulation of Fra-1 proteins in tumour cells are not well understood. Here, we show that turnover of Fra-1, which does not require ubiquitylation, is cooperatively regulated by two distinct mechanisms-association with the 19S proteasomal subunit, TBP-1, and by a C-terminal degron, which acts independently of TBP-1, but is regulated by RAS-ERK (extracellular signal-regulated kinase) signalling. TBP-1 depletion stabilized Fra-1 and further increased its levels in tumour cells expressing RAS-ERK pathway oncogenes. These effects correlated with increased AP-1 transcriptional activity. We suggest that during Fra-1 degradation, association with TBP-1 provides a mechanism for ubiquitin-independent proteasomal recognition, while the C terminus of the protein regulates its subsequent proteolytic processing.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , ATPases Associated with Diverse Cellular Activities , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Signal Transduction , Transcription Factor AP-1/metabolism , ras Proteins/metabolism
11.
Oncogene ; 29(7): 992-1002, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-19935714

ABSTRACT

Inhibition of Notch signaling is effective in inhibiting colon tumorigenesis, but targeting specific components of the pathway may provide more effective strategies. Here we show that the expression of Jagged1, a ligand for canonical Notch signaling, was restricted to enteroendocrine cells or undetectable in the mucosa of the human small and large intestine, respectively. In contrast, increased expression characterized half of human colon tumors, although not all tumors with elevated Wnt signaling displayed elevated Jagged1. Increased Jagged1 was also present in intestinal tumors of Apc(1638N/+) and Apc(Min/+) mice, but to a higher level and more frequently in the former, and in 90% of mouse tumors Notch signaling was elevated when Jagged1 was elevated. In the human HT29Cl16E colonic carcinoma cell line, induction of goblet cell differentiation by contact inhibition of growth depended on the loss of Jagged1-mediated Notch activation, with signaling through Notch1 and Notch2 acting redundantly. Therefore, targeting of Jagged1 could be effective in downregulating Notch signaling in a subset of tumors, but may avoid the limiting gastrointestinal toxicity caused by pharmacological inhibition of Notch signaling.


Subject(s)
Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Membrane Proteins/genetics , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Line, Tumor , Humans , Immunohistochemistry , Intestinal Neoplasms/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Intestine, Small/pathology , Jagged-1 Protein , Mice , Serrate-Jagged Proteins
13.
Am J Physiol ; 272(4 Pt 1): G705-12, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9142899

ABSTRACT

Control of paracellular permeability in the colonic epithelium is fundamental to its functional competence. This study examines the relationship between physiologically relevant short-chain fatty acids (SCFAs) and paracellular permeability using the Caco-2 cell line model. Butyrate induced a concentration-dependent, reversible increase in transepithelial resistance (TER) that was maximal after 72 h. Butyrate (2 mM) increased TER by 299 +/- 69% (mean +/- SE; n = 5; P < 0.05; t-test) and reduced mannitol flux to 52 +/- 11% (P < 0.05) of control. The effect of butyrate was dependent on protein synthesis and gene transcription but not dependent on its oxidation or activation of adenosine 3',5'-cyclic monophosphate. The other SCFAs, propionate and acetate, also induced a concentration-dependent increase in TER. The effect of butyrate paralleled changes in cellular differentiation, because alkaline phosphatase activity, carcinoembryonic antigen expression, and dome formation were increased. Furthermore, other differentiating agents (dimethyl sulfoxide and retinoic acid) also increased TER. Thus SCFAs reduce paracellular permeability in the Caco-2 cell line, possibly by promotion of a more differentiated phenotype. If such an effect occurs in vivo, it may have ramifications for the biology and pathobiology of colonic mucosa.


Subject(s)
Acetic Acid/pharmacology , Butyrates/pharmacology , Fatty Acids, Volatile/pharmacology , Intestinal Mucosa/metabolism , Propionates/pharmacology , Butyric Acid , Caco-2 Cells , Cell Differentiation/drug effects , Cell Division/drug effects , Electric Impedance , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Mannitol/pharmacokinetics , Permeability/drug effects
14.
Gut ; 44(3): 394-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10026327

ABSTRACT

BACKGROUND: Dietary fibre influences the turnover and differentiation of the colonic epithelium, but its effects on barrier function are unknown. AIMS: To determine whether altering the type and amount of fibre in the diet affects paracellular permeability of intestinal epithelium, and to identify the mechanisms of action. METHODS: Rats were fed isoenergetic low fibre diets with or without supplements of wheat bran (10%) or methylcellulose (10%), for four weeks. Paracellular permeability was determined by measurement of conductance and 51Cr-EDTA flux across tissue mounted in Ussing chambers. Faecal short chain fatty acid (SCFA) concentrations were assessed by gas chromatography, epithelial kinetics stathmokinetically, and mucosal brush border hydrolase activities spectrophotometrically. RESULTS: Body weight was similar across the dietary groups. Conductance and 51Cr-EDTA flux were approximately 25% higher in animals fed no fibre, compared with those fed wheat bran or methylcellulose in the distal colon, but not in the caecum or jejunum. Histologically, there was no evidence of epithelial injury or erosion associated with any diet. The fibres exerted different spectra of effects on luminal SCFA concentrations and pH, and on mucosal indexes, but both bulked the faeces, were trophic to the epithelium, and stimulated expression of a marker of epithelial differentiation. CONCLUSIONS: Both a fermentable and a non-fermentable fibre reduce paracellular permeability specifically in the distal colon, possibly by promoting epithelial cell differentiation. The mechanisms by which the two fibres exert their effects are likely to be different.


Subject(s)
Cell Membrane Permeability/physiology , Colon/physiology , Dietary Fiber/administration & dosage , Animals , Colon/cytology , Conductometry , Dietary Fiber/classification , Edetic Acid/pharmacokinetics , Epithelium , Fatty Acids, Volatile/pharmacokinetics , Male , Rats , Rats, Sprague-Dawley
15.
Gastroenterology ; 120(4): 889-99, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231943

ABSTRACT

BACKGROUND & AIMS: The short-chain fatty acid butyrate induces cell cycle arrest, differentiation, and apoptosis in colon cancer cells, but often induces opposite effects in normal colonic epithelial cells. We determined whether response to butyrate is dependent on the basal differentiation status of colonic epithelial cells. METHODS: Caco-2 cells at progressive stages of differentiation were treated with butyrate, and endpoints were measured. RESULTS: Response of Caco-2 cells to butyrate was dependent on their differentiation status. Butyrate maximally stimulated cell cycle arrest, apoptosis, alkaline phosphatase activity, transepithelial resistance, cell migration, urokinase receptor expression, and interleukin 8 secretion in undifferentiated Caco-2 cells, whereas differentiated Caco-2 cells were essentially resistant to these effects. Consistently, butyrate selectively induced histone hyperacetylation in undifferentiated Caco-2 cells. This resistance was also observed during HT29cl.19A cell differentiation, but not in the nondifferentiating SW620 cell line. Finally, the rate of butyrate use significantly increased as Caco-2 cells underwent spontaneous differentiation. CONCLUSIONS: Colonic epithelial cells become progressively more refractory to the effects of butyrate during absorptive cell differentiation. We postulate that this resistance is caused by the rapid use of butyrate by differentiated Caco-2 cells, which likely results in low intracellular concentrations and subsequently in its inability to inhibit histone deacetylase.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Colon/cytology , Intestinal Mucosa/cytology , Acetylation , Alkaline Phosphatase/metabolism , Caco-2 Cells , Cell Differentiation/drug effects , Cell Line , Cell Movement/physiology , Colon/metabolism , Colon/physiology , Drug Resistance , Electric Impedance , Histones/metabolism , Humans , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator
16.
J Gastroenterol Hepatol ; 14(9): 873-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10535468

ABSTRACT

BACKGROUND AND AIMS: The effects of butyrate on colonic epithelial barrier function are poorly understood. The aim of this study was to examine the short-term effects of butyrate on paracellular permeability of rat distal colonic epithelium. METHODS: Mucosa mounted in Ussing chambers was treated with butyrate (1-10 mmol/L) for 4 h. Transepithelial conductance, [51Cr]-EDTA flux, mucosal brush border hydrolase activity and epithelial kinetics, using proliferating cell nuclear antigen (PCNA) staining, were measured. RESULTS: On exposure to butyrate (10 mmol/L, but not 1 or 5 mmol/L), transepithelial conductance was 65 +/- 2% higher (mean +/- SEM; n = 8, P < 0.05, paired t-test) and the rate coefficient for [51Cr]-EDTA flux was 65 +/- 25% higher (P = 0.03) than those of control tissue. Histologically, the epithelium exhibited no signs of injury, but butyrate-treated tissue exhibited interstitial oedema consistent with water uptake in association with butyrate absorption. Butyrate caused a reduction in crypt column height to 30.6 +/- 1.6 cells from 33.4 +/- 1.8 cells in controls (n = 10, P < 0.03), but the number of cells per crypt column staining with PCNA was unchanged. Butyrate significantly reduced the mucosal activities of alkaline phosphatase by 40 +/- 16%, maltase by 54 +/- 12% and dipeptidyl peptidase IV by 41 +/- 14%. CONCLUSIONS: Acute exposure to butyrate increased paracellular permeability in rat distal colon. The mechanism involved may relate to the loss of differentiated surface epithelial cells, or as a physiological response to Na+-coupled butyrate uptake.


Subject(s)
Butyrates/pharmacology , Colon/drug effects , Intestinal Mucosa/drug effects , Permeability/drug effects , Alkaline Phosphatase/metabolism , Animals , Cell Cycle , Cell Differentiation , Colon/cytology , Colon/metabolism , Dipeptidyl Peptidase 4/metabolism , Electrophysiology , Hydrolases/metabolism , In Vitro Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Microvilli , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric
17.
Cell Growth Differ ; 10(10): 713-20, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10547075

ABSTRACT

Short-chain fatty acids play a critical role in colonic homeostasis because they stimulate pathways of growth arrest, differentiation, and apoptosis. These effects have been well characterized in colonic cell lines in vitro. We investigated the role of beta-catenin-Tcf signaling in these responses to butyrate and other well-characterized inducers of apoptosis of colonic epithelial cells. Unlike wild-type APC, which down-regulates Tcf activity, butyrate, as well as sulindac and trichostatin A, all inducers of G0-G1 cell cycle arrest and apoptosis in the SW620 colonic carcinoma cell line, up-regulate Tcf activity. In contrast, structural analogues of butyrate that do not induce cell cycle arrest or apoptosis and curcumin, which stimulates G2-M arrest without inducing apoptosis, do not alter Tcf activity. Similar to the cell cycle arrest and apoptotic cascade induced by butyrate, the up-regulation of Tcf activity is dependent upon the presence of a mitochondrial membrane potential, unlike the APC-induced down-regulation, which is insensitive to collapse of the mitochondrial membrane potential. Moreover, the butyrate-induced increase in Tcf activity, which is reflected in an increase in beta-catenin-Tcf complex formation, is independent of the down-regulation caused by expression of wild-type APC. Thus, butyrate and wild-type APC have different and independent effects on beta-catenin-Tcf signaling. These data are consistent with other reports that suggest that the absence of wild-type APC, associated with the up-regulation of this signaling pathway, is linked to the probability of a colonic epithelial cell entering an apoptotic cascade.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Butyrates/pharmacology , Colonic Neoplasms/pathology , Curcumin/pharmacology , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Sulindac/pharmacology , Trans-Activators , Transcription Factors/metabolism , Animals , G2 Phase , Humans , Lymphoid Enhancer-Binding Factor 1 , Mice , Up-Regulation , Valinomycin/pharmacology , beta Catenin
18.
J Cell Physiol ; 183(3): 347-54, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10797309

ABSTRACT

Caco-2 cells differentiate spontaneously when cultured in confluence and on exposure to the physiologically relevant short-chain fatty acid, butyrate. This study aimed to compare the phenotype induced by these pathways and their relations to cell turnover. Caco-2 cells were treated with butyrate at a nontoxic concentration of 2 mM for 3 days, or allowed to spontaneously differentiate for 0-21 days. Brush border hydrolase activities and carcinoembryonic antigen (CEA) expression, transepithelial resistance and dome formation, expression of components of the urokinase system, and cell turnover by flow cytometry, and the degree of DNA fragmentation were quantified. Butyrate induced increases in alkaline phosphatase activity and CEA expression but not the activities of other hydrolases, while culture alone induced progressive increases in the activities/expression of all markers. Butyrate induced a significantly greater increase in transepithelial resistance (TER) than occurred during culture alone but the densities of domes were similar. Butyrate induced a ninefold increase in urokinase receptor expression and twofold increase in urokinase activity, while culture alone induced a significantly smaller increase in receptor expression, an increase in plasminogen activator inhibitor-1 but no change in activity. While both stimuli induced cell cycle arrest, only butyrate increased the proportion of cells undergoing apoptosis. In conclusion, differentiation of Caco-2 cells can proceed along multiple pathways but does not necessarily lead to apoptosis. The phenotypic changes during spontaneous differentiation mimic those that occur in normal colonic epithelial cells in vivo during their migration from the crypt base to neck, while butyrate-induced effects more closely follow those occurring when normal colonic epithelial cells migrate from crypt neck to the surface compartment.


Subject(s)
Apoptosis/physiology , Butyrates/pharmacology , Cell Differentiation/physiology , Alkaline Phosphatase/metabolism , Apoptosis/drug effects , Caco-2 Cells , Carcinoembryonic Antigen/metabolism , Cell Cycle , Cell Differentiation/drug effects , DNA Fragmentation , Flow Cytometry , Humans , Hydrolases/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Kinetics , Microvilli/enzymology , Phenotype , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Urokinase-Type Plasminogen Activator/metabolism
19.
J Cell Physiol ; 182(2): 222-31, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10623886

ABSTRACT

LIM1215 colon cancer cells were used as a model of human colonic epithelium to examine the effects of butyrate on protein kinase C (PKC) activity and isoform expression. On Western blot analysis, LIM1215 cells express the PKC isoforms alpha, beta, epsilon, zeta, and lambda, but not gamma, straight theta, or micro. Treatment with 2 mM butyrate for 48 h reduced cellular PKC activity up to 50% and specifically reduced the expression of PKC alpha and PKC epsilon. Similar results were obtained using Caco-2 colon cancer cells. These effects were neither a consequence of the induction of differentiation itself nor the result of direct or indirect activation of PKC. Although dependent on gene transcription and protein synthesis, the effect was not due to a reduction in the synthesis of PKC protein. Butyrate's effect was independent of its beta-oxidation but was mimicked, at least in part, by trichostatin A, an inhibitor of histone deacetylase.


Subject(s)
Colon/enzymology , Fatty Acids, Volatile/physiology , Intestinal Mucosa/enzymology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Butyrates/pharmacology , Cell Differentiation/physiology , Cell Line , Colon/cytology , Enzyme Activation , Histone Deacetylase Inhibitors , Humans , Intestinal Mucosa/cytology , Isoenzymes/drug effects , Protein Kinase C/drug effects , Protein Synthesis Inhibitors/pharmacology , Transcription, Genetic/physiology
20.
Carcinogenesis ; 20(4): 539-44, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10223179

ABSTRACT

Butyrate may have paradoxical effects on epithelial cells of similar origin. This study aimed to examine the hypothesis that one mechanism that dictates a cell's response to butyrate is its state of activation. First, the responses to 24 h exposure to butyrate (1-2 mM) of normal and neoplastic human colonic epithelial cells activated by their isolation and primary culture, and of colon cancer cell lines, LIM1215 and Caco-2, were examined. In primary cultures of normal and cancer cells, butyrate had no effect on alkaline phosphatase activities but significantly suppressed urokinase receptor expression by a mean +/- SEM of 30 +/- 12% and 36 +/- 9%, respectively. Interleukin-8 secretion was suppressed by 44 +/- 7% in normal cells (P < 0.05) but was unchanged in cancer cells. In contrast, the cell lines significantly increased alkaline phosphatase activities by >50%, urokinase receptor expression >2-fold and interleukin-8 secretion >3-fold in response to butyrate. Secondly, the effect of butyrate on Caco-2 cells was examined with or without prior exposure to a specific activating stimulus [tumour necrosis factor alpha (TNF alpha)]. Interleukin-8 secretion increased by 145 +/- 23% and 132 +/- 17% on 24 h exposure to 2 mM butyrate or 0.1 microM TNF alpha alone, respectively. However, in cells pre-treated with TNF alpha, butyrate significantly inhibited secretion by 34 +/- 7% below unstimulated levels. The response to butyrate of urokinase receptor, whose expression was not stimulated by TNF alpha, was unchanged. These effects were mimicked by trichostatin A, an inhibitor of histone deacetylase, suggesting that butyrate's paradoxical effects may have been operating by the same mechanism. In conclusion, some of the paradoxical effects of butyrate do not appear to represent inherent differences between normal and transformed cells. Rather, the response may be determined by the state of activation of the cells.


Subject(s)
Butyrates/pharmacology , Colon/drug effects , Intestinal Mucosa/drug effects , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Colon/cytology , Colonic Neoplasms/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Interleukin-8/metabolism , Intestinal Mucosa/cytology , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL