Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Integr Biol (Camb) ; 9(12): 979-987, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29186222

ABSTRACT

We explored the use of irreversible electroporation (IRE) and high-frequency irreversible electroporation (H-FIRE) to induce cell death of tumor-initiating cells using a mouse ovarian surface epithelial (MOSE) cancer model. Tumor-initiating cells (TICs) can be successfully destroyed using pulsed electric field parameters common to irreversible electroporation protocols. Additionally, high-frequency pulses seem to induce cell death of TICs at significantly lower electric fields suggesting H-FIRE can be used to selectively target TICs and malignant late-stage cells while sparing the non-malignant cells in the surrounding tissue. We evaluate the relationship between threshold for cell death from H-FIRE pulses and the capacitance of cells as well as other properties that may play a role on the differences in the response to conventional IRE versus H-FIRE treatment protocols.


Subject(s)
Electroporation , Neoplasms, Glandular and Epithelial/therapy , Neoplastic Stem Cells/cytology , Ovarian Neoplasms/therapy , Animals , Carcinoma, Ovarian Epithelial , Cell Death , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Size , Collagen Type I/metabolism , Disease Models, Animal , Electrodes , Electromagnetic Fields , Female , Mice , Mice, Inbred C57BL , Models, Theoretical
2.
Clin Cancer Res ; 7(12): 4280-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751530

ABSTRACT

PURPOSE: Epithelial ovarian cancer has no reliable marker for early detection and no known specific premalignant changes. Human ovarian surface epithelial (HOSE) cells expressing human papillomavirus type 16 (HPV-16) E6/E7 genes undergo crisis, and surviving cells exhibit an immortalized phenotype. Cells show an increasingly invasive phenotype on collagen rafts over time. To ascertain the nature of this aberrant growth, we characterized this spontaneous progression of HOSE cells from a benign to an invasive phenotype using histopathology, immunophenotyping, and tumorigenesis assays. EXPERIMENTAL DESIGN: At various passages, cells were monitored for growth on collagen, response to tumor necrosis factor alpha and daunorubicin, immunohistochemistry and Western blot analysis of E-cadherin and beta-catenin, growth in soft agar, and tumor formation in immunodeficient mice. RESULTS: As passage number increased, cells became increasingly aggressive on collagen, with more pronounced focal stratification and invasion. Furthermore, late-passage cells were more resistant to the apoptotic effects of TNF-alpha and daunorubicin than earlier-passage cells. E-cadherin expression was limited to early-passage cells, whereas beta-catenin was expressed regardless of passage. Cells invading collagen formed colonies in soft agar at low efficiency but were not tumorigenic in immunodeficient mice. Some cultures recovered from colonies grew in soft agar at high efficiencies, and one was tumorigenic. CONCLUSIONS: HOSE cells expressing E6/E7, over time, develop characteristics of malignant cells and produce tumors consistent with an ovarian surface epithelium lineage. Progression of HOSE cells from a benign to an invasive phenotype in vitro may provide a model to dissect the progression of ovarian cancer.


Subject(s)
Cell Transformation, Neoplastic , Epithelial Cells/pathology , Oncogene Proteins, Viral/genetics , Ovary/pathology , Repressor Proteins , Trans-Activators , Cadherins/analysis , Cell Culture Techniques/methods , Cell Line , Cytoskeletal Proteins/analysis , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Papillomavirus E7 Proteins , beta Catenin
3.
Cancer Res ; 61(18): 6723-9, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559543

ABSTRACT

Sphingolipid consumption suppresses colon carcinogenesis, but the specific genetic defect(s) that can be bypassed by these dietary components are not known. Colon tumors often have defect(s) in the adenomatous polyposis coli (APC)/beta-catenin regulatory system. Therefore, C57Bl/6J(Min/+) mice with a truncated APC gene product were fed diets supplemented with ceramide, sphingomyelin, glucosylceramide, lactosylceramide, and ganglioside G(D3) (a composition similar in amount and type to that of dairy products) to determine whether tumorigenesis caused by this category of genetic defect is suppressed. Sphingolipid feeding reduced the number of tumors in all regions of the intestine, and caused a marked redistribution of beta-catenin from a diffuse (cytosolic plus membrane) pattern to a more "normal" localization at mainly intercellular junctions between intestinal epithelial cells. The major digestion product of complex sphingolipids is sphingosine, and treatment of two human colon cancer cell lines in culture (SW480 and T84) with sphingosine reduced cytosolic and nuclear beta-catenin, inhibited growth, and induced cell death. Ceramides, particularly long-chain ceramides, also had effects. Thus, dietary sphingolipids, presumably via their digestion products, bypass or correct defect(s) in the APC/beta-catenin regulatory pathway. This may be at least one mechanism whereby dietary sphingolipids inhibit colon carcinogenesis, and might have implications for dietary intervention in human familial adenomatous polyposis and colon cancer.


Subject(s)
Cytoskeletal Proteins/metabolism , Intestinal Neoplasms/prevention & control , Sphingolipids/pharmacology , Trans-Activators , Adenomatous Polyposis Coli Protein , Animals , Cattle , Cell Division/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytoskeletal Proteins/physiology , Cytosol/drug effects , Cytosol/metabolism , Diet , Humans , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Sphingolipids/administration & dosage , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/blood , Sphingosine/toxicity , Tumor Cells, Cultured , beta Catenin
4.
J Nutr ; 130(3): 522-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10702579

ABSTRACT

Dietary sphingomyelin (SM) inhibits early stages of colon cancer (appearance of aberrant crypt foci, ACF) and decreases the proportion of adenocarcinomas vs. adenomas in 1,2-dimethylhydrazine (DMH)-treated CF1 mice. To elucidate the structural specificity of this inhibition, the effects of the other major sphingolipids in milk (glycosphingolipids) were determined. Glucosylceramide (GluCer), lactosylceramide (LacCer) and ganglioside G(D3) were fed individually to DMH-treated (six doses of 30 mg/kg body weight) female CF1 mice at 0.025 or 0.1 g/100 g of the diet for 4 wk. All reduced the number of ACF by > 40% (P < 0.001), which is comparable to the reduction by SM in earlier studies. Immunohistochemical analysis of the colons revealed that sphingolipid feeding also reduced proliferation, with the most profound effect (up to 80%; P < 0.001) in the upper half of the crypts. Since the bioactive backbones of the glycosphingolipids (i.e., ceramide and other metabolites) are the likely mediators of these effects, the susceptibility of these complex sphingolipids to digestion in the colon was examined by incubating 500 microgram of each sphingolipid with colonic segments from mice and analysis of substrate disappearance and product formation by tandem mass spectrometry. All of the sphingolipids (including SM) disappeared over time with a substantial portion appearing as ceramide. Partially hydrolyzed intermediates (such as GluCer from LacCer or G(D3)) were not detected, which suggests that the cleavage involves colonic (or microflora) endoglycosidases. In summary, consumption of dairy SM and glycosphingolipids suppresses colonic cell proliferation and ACF formation in DMH-treated mice; hence, many categories of sphingolipids affect these key events in colon carcinogenesis.


Subject(s)
Adenocarcinoma/prevention & control , Adenoma/prevention & control , Colonic Neoplasms/prevention & control , Diet , Glycosphingolipids/administration & dosage , Glycosphingolipids/therapeutic use , 1,2-Dimethylhydrazine/toxicity , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Adenoma/chemically induced , Adenoma/pathology , Animals , Apoptosis/drug effects , Carcinogens/toxicity , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Fatty Acids/analysis , Female , Glycosphingolipids/analysis , Linear Models , Mice , Milk/chemistry
5.
Cancer Res ; 59(22): 5768-72, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10582697

ABSTRACT

Dietary sphingolipids inhibit chemically induced colon cancer in mice. The most likely mediators of this effect are the metabolites ceramide (Cer) and sphingosine, which induce growth arrest and apoptosis in transformed cells. Sphingolipids are digested in both the upper and the lower intestine; therefore, a more colon-specific method of delivery of sphingolipids might be useful. A Cer analogue with a D-glucuronic acid attached at the primary hydroxyl of N-palmitoyl-D-sphingosine (Cer-beta-glucuronide) was synthesized and evaluated as a substrate for Escherichia coli beta-glucuronidase and colonic digestion, as well as for suppression of early events in colon carcinogenesis in CFI mice treated with 1,2-dimethylhydrazine. Purified beta-glucuronidase (EC 3.2.1.31) and colonic segments (as a source of colonic enzymes and microflora) hydrolyzed Cer-beta-glucuronide to release Cer, as analyzed by tandem mass spectrometry. More than 75% of the Cer-beta-glucuronide was cleaved in an 8-h incubation with the colonic segments. When Cer-beta-glucuronide was administered for 4 weeks as 0.025% and 0.1% of the diet (AIN 76A) to 1,2-dimethylhydrazine-treated mice, there were significant reductions in colonic cell proliferation, as determined by in vivo BrdUrd incorporation, and in the appearance of aberrant crypt foci. The effect of dietary Cer-beta-glucuronide on aberrant crypt foci correlated significantly with the length of the colon, which suggests that Cer-beta-glucuronide was most effective when there was a larger compartment for digestion. Thus, synthetic sphingolipids that target the colon for the release of the bioactive backbones offer a promising approach to colon cancer prevention.


Subject(s)
Anticarcinogenic Agents/metabolism , Bacterial Proteins/metabolism , Colonic Neoplasms/prevention & control , Glucosylceramides/metabolism , Glucuronates/metabolism , Glucuronidase/metabolism , Precancerous Conditions/prevention & control , 1,2-Dimethylhydrazine , Animals , Anticarcinogenic Agents/chemical synthesis , Anticarcinogenic Agents/therapeutic use , Carcinogens , Ceramides/metabolism , Colon/microbiology , Colonic Neoplasms/chemically induced , Drug Screening Assays, Antitumor , Escherichia coli/enzymology , Female , Glucosylceramides/chemical synthesis , Glucosylceramides/therapeutic use , Glucuronates/chemical synthesis , Glucuronates/therapeutic use , Hydrolysis , Mice , Precancerous Conditions/chemically induced , Weight Gain/drug effects
6.
J Nutr ; 129(7): 1239-50, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10395583

ABSTRACT

Eukaryotic organisms as well as some prokaryotes and viruses contain sphingolipids, which are defined by a common structural feature, i.e. , a "sphingoid base" backbone such as D-erythro-1,3-dihydroxy, 2-aminooctadec-4-ene (sphingosine). The sphingolipids of mammalian tissues, lipoproteins, and milk include ceramides, sphingomyelins, cerebrosides, gangliosides and sulfatides; plants, fungi and yeast have mainly cerebrosides and phosphoinositides. The total amounts of sphingolipids in food vary considerably, from a few micromoles per kilogram (fruits) to several millimoles per kilogram in rich sources such as dairy products, eggs and soybeans. With the use of the limited data available, per capita sphingolipid consumption in the United States can be estimated to be on the order of 150-180 mmol (approximately 115-140 g) per year, or 0.3-0.4 g/d. There is no known nutritional requirement for sphingolipids; nonetheless, they are hydrolyzed throughout the gastrointestinal tract to the same categories of metabolites (ceramides and sphingoid bases) that are used by cells to regulate growth, differentiation, apoptosis and other cellular functions. Studies with experimental animals have shown that feeding sphingolipids inhibits colon carcinogenesis, reduces serum LDL cholesterol and elevates HDL, suggesting that sphingolipids represent a "functional" constituent of food. Sphingolipid metabolism can also be modified by constituents of the diet, such as cholesterol, fatty acids and mycotoxins (fumonisins), with consequences for cell regulation and disease. Additional associations among diet, sphingolipids and health are certain to emerge as more is learned about these compounds.


Subject(s)
Food Analysis , Nutritional Physiological Phenomena , Sphingolipids/physiology , Cell Division/drug effects , Colonic Neoplasms/prevention & control , Diet , Digestion/physiology , Humans , Sphingolipids/analysis , Sphingolipids/metabolism , Sphingolipids/therapeutic use
7.
Chem Phys Lipids ; 102(1-2): 131-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-11001567

ABSTRACT

Sphingolipids modulate many aspects of cell function, including the expression of cytochrome P450, a superfamily of heme proteins that participate in the oxidation of a wide range of compounds of both endogenous (steroid hormones and other lipids) and exogenous (e.g. alcohol, drugs and environmental pollutants) origin. Cytochrome P450-2C11 (CYP 2C11) is down-regulated in response to interleukin-1beta (IL-1beta), and this response involves the hydrolysis of sphingomyelin to ceramide as well as ceramide to sphingosine, and phosphorylation of sphingosine to sphingosine 1-phosphate. Activation of ceramidase(s) are a key determinant of which bioactive sphingolipid metabolites are formed in response to IL-1beta. Ceramidase activation also appears to account for the loss of expression of CYP 2C11 when hepatocytes are placed in cell culture, and the restoration of expression when they are plated on Matrigel; hence, this pathway is influenced by, and may mediate, interactions between hepatocytes and the extracellular matrix. Recent studies using inhibitors of sphingolipid metabolism have discovered that sphingolipids are also required for the induction of CYP1A1 by 3-methylcholanthrene, however, in this case, the requirement is for de novo sphingolipid biosynthesis rather than the turnover of complex sphingolipids. These findings illustrate how changes in sphingolipid metabolism can influence the regulation of at least several isoforms of cytochrome P450.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Sphingolipids/physiology , Steroid 16-alpha-Hydroxylase , Animals , Humans , Isoenzymes/biosynthesis , Sphingolipids/metabolism , Steroid Hydroxylases/biosynthesis
9.
J Biol Chem ; 273(30): 19060-4, 1998 Jul 24.
Article in English | MEDLINE | ID: mdl-9668088

ABSTRACT

Fumonisin B1 (FB1) is the predominant member of a family of mycotoxins produced by Fusarium moniliforme (Sheldon) and related fungi. Certain foods also contain the aminopentol backbone (AP1) that is formed upon base hydrolysis of the ester-linked tricarballylic acids of FB1. Both FB1 and, to a lesser extent, AP1 inhibit ceramide synthase due to structural similarities between fumonisins (as 1-deoxy-analogs of sphinganine) and sphingoid bases. To explore these structure-function relationships further, erythro- and threo-2-amino, 3-hydroxy- (and 3, 5-dihydroxy-) octadecanes were prepared by highly stereoselective syntheses. All of these analogs inhibit the acylation of sphingoid bases by ceramide synthase, and are themselves acylated with Vmax/Km of 40-125 for the erythro-isomers (compared with approximately 250 for D-erythro-sphinganine) and 4-6 for the threo-isomers. Ceramide synthase also acylates AP1 (but not FB1, under the conditions tested) to N-palmitoyl-AP1 (PAP1) with a Vmax/Km of approximately 1. The toxicity of PAP1 was evaluated using HT29 cells, a human colonic cell line. PAP1 was at least 10 times more toxic than FB1 or AP1 and caused sphinganine accumulation as an inhibitor of ceramide synthase. These studies demonstrate that: the 1-hydroxyl group is not required for sphingoid bases to be acylated; both erythro- and threo-isomers are acylated with the highest apparent Vmax/Km for the erythro-analogs; and AP1 is acylated to PAP1, a new category of ceramide synthase inhibitor as well as a toxic metabolite that may play a role in the diseases caused by fumonisins.


Subject(s)
Carboxylic Acids/metabolism , Fumonisins , Mycotoxins/metabolism , Oxidoreductases/metabolism , Palmitic Acid/metabolism , Sphingosine/analogs & derivatives , Acylation , Carboxylic Acids/chemistry , Humans , Hydrolysis , Kinetics , Mycotoxins/chemistry , Oxidoreductases/antagonists & inhibitors , Pancreatitis-Associated Proteins , Sphingosine/metabolism
10.
Toxicol Appl Pharmacol ; 148(2): 252-60, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9473533

ABSTRACT

Fumonisin B1 (FB1) and aminopentol (AP1) (which is formed by hydrolysis of FB1) are found in corn contaminated with some strains of Fusarium moniliforme. Incubation of HT29 cells (a human colonic cell line) with FB1 or AP1 caused a significant reduction in cell number; AP1 was less potent, with 50 microM AP1 causing the same reduction (ca. 30% after 24 h) as 10 microM FB1. The reduction in cell number reflected increases in DNA fragmentation and the percentage of apoptotic cells. Both FB1 and AP1 caused the accumulation of sphinganine (25- and 35-fold by 10 microM FB1 and 50 microM AP1, respectively); thus, concentrations of FB1 and AP1 that caused comparable reductions in cell number were also similar with respect to elevation of sphinganine, a compound that is growth inhibitory and cytotoxic. Inhibition of the first step of sphingolipid biosynthesis with ISP-1 prevented the elevation in sphinganine, DNA fragmentation, and apoptosis induced by FB1. Therefore, these effects of FB1 on HT29 cells can be attributed to the accumulation of sphinganine. Since consumption of food contaminated with Fusarium moniliforme (Sheldon) exposes colonic cells to these mycotoxins, the possibility that FB1 and AP1 are toxic for intestinal cells in vivo should be evaluated, especially in the light of the recent report (Bhat et al., Clin. Toxicol. 35, 249, 1997) describing intestinal disturbances in humans after consumption of moldy corn and sorghum containing fumonisins.


Subject(s)
Apoptosis , Carboxylic Acids/toxicity , Carcinogens, Environmental/toxicity , Fumonisins , HT29 Cells/drug effects , Mycotoxins/toxicity , Antifungal Agents/pharmacology , Carboxylic Acids/antagonists & inhibitors , Carboxylic Acids/metabolism , Cell Count/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Enzyme Inhibitors/metabolism , Fatty Acids, Monounsaturated/pharmacology , Food Contamination , Humans , Sphingolipids/analysis , Sphingolipids/biosynthesis , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Zea mays
11.
J Nutr ; 127(5 Suppl): 830S-833S, 1997 05.
Article in English | MEDLINE | ID: mdl-9164247

ABSTRACT

Sphingolipids are highly bioactive compounds that participate in the regulation of cell growth, differentiation, diverse cell functions, and apoptosis. They are present in both plant and animal foods in appreciable amounts, but little is known about their nutritional significance. Recent studies have shown that feeding sphingomyelin to female CF1 mice treated with a colon carcinogen (1,2-dimethylhydrazine) reduced the number of aberrant colonic crypt foci; longer-term feeding also affected the appearance of colonic adenocarcinomas. Therefore, dietary sphingolipids should be considered in studies of the relationships between diet and cancer. Sphingolipids have also surfaced as important factors in understanding the mechanism of action of a recently discovered family of mycotoxins, termed fumonisins. Fumonisins are produced by fungi commonly found on maize and a few related foods, and their consumption can result in equine leukoencephalomalacia, porcine pulmonary edema and a number of other diseases of veterinary animals and, perhaps, humans. A cellular target of fumonisins is the enzyme ceramide synthase, and disruption of sphingolipid metabolism by fumonisins has been established by studies with both cells in culture and animals that have consumed these toxic mycotoxins. These findings underscore the ways in which sphingolipids and agents that affect sphingolipid utilization should be given consideration in selecting animal diets for nutritional and toxicological studies.


Subject(s)
Animal Feed , Diet , Sphingolipids/administration & dosage , Sphingolipids/antagonists & inhibitors , Animals , Colonic Neoplasms/prevention & control , Mycotoxins/pharmacology , Sphingolipids/physiology
12.
Nutr Cancer ; 28(1): 81-5, 1997.
Article in English | MEDLINE | ID: mdl-9200154

ABSTRACT

Supplementation of the diet of CF1 mice with sphingomyelin isolated from milk has been shown to reduce the number of aberrant crypt foci (ACF) and the appearance of colonic adenocarcinoma induced by 1,2-dimethylhydrazine (Schmelz et al., Cancer Res 56, 4936-4941, 1996). The objective of this study was to determine whether chemically synthesized sphingomyelin reduces the appearance of ACF, one of the earliest morphological changes in the development of colonic tumors, and to investigate the specificity of this inhibition for the unsaturated sphingoid base backbone. 1,2-Dimethylhydrazine was administered intraperitoneally to female CF1 mice, then the animals were fed a semipurified AIN 76A diet without supplementation (controls) or supplemented with 0.1% (wt/wt) sphingomyelin isolated from skim milk powder, synthetic N-palmitoylsphingomyelin, or N-palmitoyldihydrosphingomyelin for four weeks. The number of ACF in the sphingomyelin-fed groups was significantly lower than in the control by 54% (p = 0.002), 52% (p = 0.002), and 70% (p < 0.0001) for milk sphingomyelin, synthetic sphingomyelin, and synthetic dihydrosphingomyelin, respectively. Suppression of ACF by the synthetic dihydrosphingomyelin was significantly greater than by synthetic sphingomyelin (p = 0.035). These findings establish that sphingomyelin, and not merely a possible contaminant of the naturally occurring sphingomyelin preparation used previously, suppresses ACF formation. Furthermore, the greater potency of dihydrosphingomyelin reveals that the 4,5-trans double bond of the sphingoid backbone is not required for this suppression.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/prevention & control , Precancerous Conditions/prevention & control , Sphingolipids/chemistry , Sphingomyelins/chemistry , Sphingomyelins/therapeutic use , 1,2-Dimethylhydrazine , Adenocarcinoma/prevention & control , Animals , Diet , Dimethylhydrazines , Female , Mice , Milk/chemistry , Precancerous Conditions/chemically induced , Sphingomyelins/administration & dosage , Weight Gain
13.
Toxicol Appl Pharmacol ; 142(1): 208-25, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9007051

ABSTRACT

The "sphingosin" backbone of sphingolipids was so named by J. L. W. Thudichum in 1884 for its enigmatic ("Sphinx-like") properties. Although still an elusive class of lipids, research on the involvement of sphingolipids in the signal transduction pathways that mediate cell growth, differentiation, multiple cell functions, and cell death has been rapidly expanding our understanding of these compounds. In addition to the newly discovered role of ceramide as an intracellular second messenger for tumor necrosis factor-alpha, IL-1beta, and other cytokines, sphingosine, sphingosine-1-phosphate, and other sphingolipid metabolites have recently been demonstrated to modulate cellular calcium homeostasis and cell proliferation. Perturbation of sphingolipid metabolism using synthetic and naturally occurring inhibitors of key enzymes of the biosynthetic pathways is aiding the characterization of these processes; for examples, inhibition of cerebroside synthase has indicated a role for ceramide in cellular stress responses including heat shock, and inhibition of ceramide synthase (by fumonisins) has revealed the role of disruption of sphingolipid metabolism in several animal diseases. Fumonisins are currently the focus of a FDA long-term tumor study. This review summarizes recent research on (i) the role of sphingolipids as important components of the diet, (ii) the role of sphingoid base metabolites and the ceramide cycle in expression of genes regulating cell growth, differentiation, and apoptosis, (iii) the use of cerebroside synthase inhibitors as tools for understanding the role of sphingolipids as mediators of cell cycle progression, renal disease, and stress responses, and (iv) the involvement of disrupted sphingolipid metabolism in animal disease and cellular deregulation associated with exposure to inhibitors of ceramide synthase and serine palmitoyltransferase, key enzymes in de novo sphingolipid biosynthesis. These findings illustrate how an understanding of the function of sphingolipids can help solve questions in toxicology and this is undoubtedly only the beginning of this story.


Subject(s)
Sphingolipids/physiology , Amidohydrolases/antagonists & inhibitors , Animal Feed , Animals , Calcium/metabolism , Carboxylic Acids/toxicity , Cell Cycle , Cell Division , Ceramidases , Ceramides/physiology , Dietary Fats , Enzyme Inhibitors/pharmacology , Food Contamination , Gene Expression Regulation/physiology , Growth Substances/physiology , Homeostasis , Humans , Mammals/metabolism , Membrane Lipids/chemistry , Membrane Lipids/physiology , Models, Biological , Morpholines/pharmacology , Mycotoxins/toxicity , Second Messenger Systems , Sphingolipids/chemistry , Stress, Physiological/metabolism
14.
Cancer Res ; 56(21): 4936-41, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8895747

ABSTRACT

Sphingolipids are hydrolyzed in the gastrointestinal tract to ceramide, sphingosine, and other metabolites that can modulate cell growth, differentiation, and apoptosis. To characterize the effects of dietary sphingolipids on colon carcinogenesis, female CF1 mice were administered 1,2-dimethylhydrazine and then fed an essentially sphingolipid-free diet supplemented with 0 to 0.1% (w/w) sphingomyelin (SM) purified from milk. As was found in a previous pilot study (D. L. Dillehay et al., J. Nutr., 124: 615-620, 1994), SM (@ 0.1%) reduced the number of aberrant colonic crypt foci (by 70%, P < 0.001) and aberrant crypts per focus (by 30%, P < 0.003), which are early indicators of colon carcinogenesis. In longer term studies, SM had no effect on colon tumor incidence or multiplicity; however, up to 31% of the tumors of mice fed SM were adenomas, whereas all of the tumors of mice fed the diet without SM were adenocarcinomas. These findings demonstrate that milk SM suppresses the appearance of more advanced, malignant tumors as well as early markers of colon carcinogenesis. Although the sphingolipid content of foods has not been widely studied, several foods (e.g., milk and soybeans) contain the sphingolipid levels used in these investigations; therefore, this class of compounds could be significant contributors to the cancer preventive effects of some foods.


Subject(s)
Adenocarcinoma/prevention & control , Adenoma/prevention & control , Colonic Neoplasms/prevention & control , Precancerous Conditions/prevention & control , Sphingomyelins/administration & dosage , 1,2-Dimethylhydrazine , Animals , Dimethylhydrazines , Female , Mice , Sphingomyelins/analysis , Sphingomyelins/metabolism
15.
J Nutr ; 125(6 Suppl): 1677S-1682S, 1995 06.
Article in English | MEDLINE | ID: mdl-7782925

ABSTRACT

Sphingolipids are found in all eukaryotic and some prokaryotic organisms and participate in the regulation of cell growth, differentiation, and diverse cell functions including cell-cell communication, cell-substratum interactions and intracellular signal transduction. Nonetheless, the field of nutrition has given scant attention to these compounds so that little is known about the following fundamental questions: What is the fate of sphingolipids that are consumed in food? Does consumption of dietary sphingolipids affect the behavior of cells in the gastrointestinal tract or other organs? How do other factors in the diet affect sphingolipid metabolism? Several recent findings underscore the importance of these questions, for examples: 1) Sphingolipids are digested throughout the GI tract to ceramide and sphingosine, which are highly bioactive compounds that affect cellular regulatory pathways; 2) addition of sphingomyelin to a standard AIN diet (which is essentially devoid of sphingolipids) reduces the appearance of aberrant colonic crypts, and perhaps the number of tumors, in mice treated with a colon carcinogen; and 3) an enzyme of sphingolipid metabolism has been discovered to be the target of a class of toxic and carcinogenic mycotoxins called fumonisins. Given these recent findings, it is possible that some of the confusion that has arisen regarding the relationships between dietary fat and disease might be due to the lack of consideration of the sphingolipids that are also present.


Subject(s)
Diet , Fumonisins , Neoplasms/prevention & control , Sphingolipids/metabolism , Animals , Carcinogens, Environmental/toxicity , Digestion , Humans , Mycotoxins/toxicity , Neoplasms, Experimental/prevention & control
16.
J Nutr ; 124(5): 615-20, 1994 May.
Article in English | MEDLINE | ID: mdl-8169652

ABSTRACT

Sphingolipids are in all eukaryotic cells and modulate cell growth, differentiation, and transformation; however, little is known about the physiological effects of their consumption. Mice were fed diets supplemented with milk sphingomyelin to determine effects on colon carcinogenesis. Cancer was initiated in CF1 mice by 1,2-dimethylhydrazine. Mice were then fed AIN76A diets supplemented with 0.025 to 0.1 g sphingomyelin/100 g for 28 wk until the supply of sphingomyelin was depleted and then fed unsupplemented diet for 24 wk. Sphingomyelin did not affect weight gain. Mice fed sphingomyelin had a 20% incidence of colon tumors compared with 47% in controls (P = 0.08 for all sphingomyelin-fed mice vs. controls). Tumors were adenomas or adenocarcinomas and located in the distal third of the colon. In shorter-term studies, colonic epithelial cell proliferation was significantly greater than controls in mice fed 0.025 g sphingomyelin/100 g diet, but not in those fed higher amounts of sphingomyelin. The number of aberrant crypts was significantly lower in 1,2-dimethylhydrazine-treated mice fed 0.05 g sphingomyelin/100 g diet than in controls. These results demonstrate that consumption of sphingomyelin affects the behavior of colonic cells. Because sphingolipids are present in food, the reduction in 1,2-dimethylhydrazine-induced premalignant lesions and the incidence of colon tumors in CF1 mice implies that these compounds may be another important class of nutritional modulators of carcinogenesis.


Subject(s)
Adenocarcinoma/prevention & control , Adenoma/prevention & control , Colonic Neoplasms/prevention & control , Sphingomyelins/therapeutic use , 1,2-Dimethylhydrazine , Adenocarcinoma/chemically induced , Adenoma/chemically induced , Administration, Oral , Animals , Carcinogens/toxicity , Cell Division/drug effects , Choristoma/chemically induced , Colonic Neoplasms/chemically induced , Dimethylhydrazines/toxicity , Female , Mice , Sphingomyelins/administration & dosage , Sphingomyelins/pharmacology , Weight Gain/drug effects
17.
J Nutr ; 124(5): 702-12, 1994 May.
Article in English | MEDLINE | ID: mdl-8169662

ABSTRACT

Sphingolipids are found in all eukaryotic organisms. However, little is known about the digestion, uptake and subsequent metabolism of these constituents of food. In this study, radiolabeled sphingolipids were placed in isolated intestinal segments of female CF1 mice, and the metabolism and distribution of the radiolabel were followed. Most of the sphingomyelin was degraded to ceramide and other products in all regions of the intestine, and increasing amounts of several [3H]-labeled sphingolipids appeared in the tissues. Small amounts of the radiolabel disappeared from the intestinal loops and appeared in liver within the first 30 to 60 min implying that neither intact sphingomyelin nor its metabolites are transported very efficiently from the intestine to other organs. There were different degrees of uptake and metabolism of sphingomyelin, [4,5-3H-sphinganyl]ceramide, and [3H]sphingosine. The [3H]sphingomyelin was also administered by gavage and the appearance along the intestine measured. After 90 min, 12% was found in the cecum and colon. These results establish that some of the sphingomyelin that enters the gastrointestinal tract is hydrolyzed and taken up by the intestine, with the lipid backbone being degraded or reutilized for complex sphingolipid synthesis; however, at least a portion passes into the large intestine. The appearance of bioactive compounds throughout the gastrointestinal tract may alter the behavior of intestinal cells.


Subject(s)
Intestinal Mucosa/metabolism , Sphingolipids/metabolism , Sphingolipids/pharmacokinetics , Administration, Oral , Animals , Carbon Radioisotopes , Ceramides/metabolism , Ceramides/pharmacokinetics , Colon/metabolism , Digestion , Female , Gastric Mucosa/metabolism , Intestinal Absorption , Mice , Sphingolipids/chemistry , Sphingosine/metabolism , Sphingosine/pharmacokinetics , Tissue Distribution , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...