Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Nanobiotechnology ; 19(1): 329, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34666761

ABSTRACT

BACKGROUND: There is an obvious correlation between ulcerative colitis and colorectal cancer, and the risk of colorectal cancer in patients with ulcerative colitis is increasing. Therefore, the combination therapy of anti-inflammatory and anti-tumor drugs may show promising to inhibit colon cancer. 5-aminosalicylic acid (5-ASA) with anti-inflammatory function is effective for maintaining remission in patients with ulcerative colitis and may also reduce colorectal cancer risk. Histone deacetylase (HDAC) plays an essential role in the progression of colon cancer. Butyric acid (BA) is a kind of HDAC inhibitor and thus shows tumor suppression to colon cancer. However, the volatile and corrosive nature of BA presents challenges in practical application. In addition, its clinical application is limited due to its non-targeting ability and low bioavailability. We aimed to synthesize a novel dual-prodrug of 5-ASA and BA, referred as BBA, to synergistically inhibit colon cancer. Further, based on the fact that folate receptor (FR) is over-expressed in most solid tumors and it has been identified to be a cancer stem cell surface marker in colon cancer, we took folate as the targeting ligand and used carboxymethyl-ß-cyclodextrin (CM-ß-CD) to carry BBA and thus prepared a novel inclusion complex of BBA/FA-PEG-CM-ß-CD. RESULTS: It was found that BBA/FA-PEG-CM-ß-CD showed significant inhibition in cell proliferation against colon cancer cells SW620. It showed a pro-longed in vivo circulation and mainly accumulated in tumor tissue. More importantly, BBA/FA-PEG-CM-ß-CD gave great tumor suppression effect against nude mice bearing SW620 xenografts. CONCLUSIONS: Therefore, BBA/FA-PEG-CM-ß-CD may have clinical potential in colon cancer therapy.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms/metabolism , Drug Delivery Systems/methods , Prodrugs , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Butyric Acid/metabolism , Butyric Acid/pharmacokinetics , Butyric Acid/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclodextrins/chemistry , Folic Acid/metabolism , Male , Mesalamine/metabolism , Mesalamine/pharmacokinetics , Mesalamine/pharmacology , Mice , Mice, Nude , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology
2.
Appl Radiat Isot ; 164: 109297, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32768887

ABSTRACT

PURPOSE: The present study analyzed different protocols of administration of boronophenylalanine (BPA) and sodium butyrate (NaB) to increase the BNCT efficacy for poorly differentiated thyroid cancer (PDTC). MATERIALS AND METHODS: Nude mice implanted with human PDTC cells (WRO) were distributed into four protocols: 1) BPA; 2) BPA + ip NaB; 3) BPA + oral NaB; 4) Control. Biodistribution and histologic studies were performed. LAT (BPA transporter) isoforms gene expression was assessed by RT-PCR. RESULTS: Tumor growth delay was observed in animals of the Protocol #3 (p < 0.05). NaB (Protocol #2) increased tumor boron uptake 2-h post BPA injection (p < 0.05). On the other hand, NaB upregulated the expression of all the isoforms of the LAT transporter in vitro. Histologic studies showed a significant decrease of mitotic activity and an increase of vacuoles in tumors of Protocol #3. Neutrons alone or combined with NaB caused some tumor growth delay (p < 0.05), while in the BNCT and BNCT + NaB groups, there was a halt in tumor growth in 70 and 80% of the animals, respectively. CONCLUSIONS: Intraperitoneally administration of NaB increased boron uptake while oral administration for a longer period of time induced tumor growth delay previous to BPA administration. The use of NaB via ip would optimize the irradiation results.


Subject(s)
Boron Neutron Capture Therapy/methods , Butyric Acid/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/radiotherapy , Animals , Butyric Acid/pharmacokinetics , Cell Differentiation , Cell Line, Tumor , Combined Modality Therapy , Histone Deacetylase Inhibitors/pharmacokinetics , Humans , Mice , Thyroid Neoplasms/pathology , Tissue Distribution , Xenograft Model Antitumor Assays
3.
Poult Sci ; 97(1): 167-176, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29077956

ABSTRACT

The hypothesis was tested that butyrate presence in the digesta of distinct gastrointestinal tract (GIT) segments of broilers leads to differential effects on digesta retention time, gut morphology, and proteolytic enzymatic activities, ultimately resulting in differences in protein digestibility. A total of 320 male day-old Ross 308 broilers were randomly assigned to 5 dietary treatments: 1) control (no butyrate), 2) unprotected butyrate (main activity in the crop and gastric regions), 3) tributyrin (main activity in the small intestine), 4) fat-coated butyrate (activity in the whole GIT) and 5) unprotected butyrate combined with tributyrin, each replicated 8 times. Rapeseed meal was used in combination with a fine dietary particle size in order to challenge the digestive capacity of young broilers. Birds were dissected at 22, 23, and 24 d of age and samples of digesta at various GIT locations as well as tissues were collected. Butyrate concentration varied significantly across GIT segments depending on treatment, indicating that the dietary contrasts were successful. The apparent ileal digestibility of methionine tended to increase when butyrate and/or propionate was present in colonic and cecal contents, possibly due to modifications of GIT development and digesta transit time. Butyrate presence in the digesta of the crop, proventriculus and gizzard, on the contrary, decreased the apparent ileal digestibility of several amino acids (AA). In addition, butyrate presence beyond the gizzard elicited anorexic effect that might be attributable to changes in intestinal enteroendocrine L-cells secretory activities. The present study demonstrates that, in broilers, effects of butyrate on digestive processes are conditioned by the GIT segment wherein the molecule is present and indicates its influence on digestive function and bioavailability of AA.


Subject(s)
Butyric Acid/metabolism , Chickens/physiology , Digestion/drug effects , Gastrointestinal Tract/drug effects , Triglycerides/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena/drug effects , Animals , Biological Availability , Butyric Acid/administration & dosage , Butyric Acid/pharmacokinetics , Diet/veterinary , Dietary Supplements/analysis , Male , Triglycerides/administration & dosage , Triglycerides/pharmacokinetics
4.
J Anim Physiol Anim Nutr (Berl) ; 100(4): 637-42, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26614344

ABSTRACT

The expression of hepatic drug-metabolizing cytochrome P450 (CYP) enzymes may be affected by several nutrition-derived compounds, such as by the commonly applied feed additive butyrate, possibly leading to feed-drug interactions. The aim of this study was to provide some evidence if butyrate can alter the activity of hepatic CYPs in chickens exposed to CYP-inducing xenobiotics, monitoring for the first time the possibility of such interaction. Ross 308 chickens in the grower phase were treated with daily intracoelomal phenobarbital (PB) injection (80 mg/kg BW), applied as a non-specific CYP-inducer, simultaneously with two different doses of intra-ingluvial sodium butyrate boluses (0.25 and 1.25 g/kg BW) for 5 days. Activity of CYP2H and CYP3A subfamilies was assessed by specific enzyme assays from isolated liver microsomes. According to our results, the lower dose of orally administered butyrate significantly attenuated the PB-triggered elevation of both hepatic CYP2H and CYP3A activities, which might be in association with the partly common signalling pathways of butyrate and CYP-inducing drugs, such as that of PB. Based on these data, butyrate may take part in pharmacoepigenetic interactions with simultaneously applied drugs or other CYP-inducing xenobiotics, with possible consequences for food safety and pharmacotherapy. Butyrate was found to be capable to maintain physiological CYP activity by attenuating CYP induction, underlining the safety of butyrate application in poultry nutrition.


Subject(s)
Animal Feed/analysis , Butyric Acid/pharmacokinetics , Chickens , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Liver/enzymology , Phenobarbital/pharmacokinetics , Administration, Oral , Animals , Butyric Acid/administration & dosage , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Regulation, Enzymologic , Liver/drug effects , Male , Phenobarbital/administration & dosage
5.
J Vet Pharmacol Ther ; 37(4): 406-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24628435

ABSTRACT

Butyrate, a commonly applied feed additive in poultry nutrition, can modify the expression of certain genes, including those encoding cytochrome P450 (CYP) enzymes. In comparative in vitro and in vivo experiments, the effect of butyrate on hepatic CYP genes was examined in primary cultures of chicken hepatocytes and in liver samples of chickens collected from animals that had been given butyrate as a feed additive. Moreover, the effect of butyrate on the biotransformation of erythromycin, a marker substance for the activity of enzymes of the CYP3A family, was investigated in vitro and in vivo. Butyrate increased the expression of the avian-specific CYP2H1 both in vitro and in vivo. In contrast, the avian CYP3A37 expression was decreased in hepatocytes following butyrate exposure, but not in the in vivo model. CYP1A was suppressed by butyrate in the in vitro experiments, and overexpressed in vivo in butyrate-fed animals. The concomitant incubation of hepatocytes with butyrate and erythromycin led to an increased CYP2H1 expression and a less pronounced inhibition of CYP3A37. In in vivo pharmacokinetic experiments, butyrate-fed animals given a single i.m. injection of erythromycin, a slower absorption phase (longer T(half-abs) and delayed T(max)) but a rapid elimination phase of this marker substrate was observed. Although these measurable differences were detected in the pharmacokinetics of erythromycin, it is unlikely that a concomitant application of sodium butyrate with erythromycin or other CYP substrates will cause clinically significant feed-drug interaction in chickens.


Subject(s)
Butyric Acid/pharmacology , Butyric Acid/pharmacokinetics , Chickens/metabolism , Erythromycin/pharmacokinetics , Liver/metabolism , Animal Feed , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Biotransformation , Butyric Acid/administration & dosage , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Diet/veterinary , Drug Interactions , Erythromycin/administration & dosage , Female , Gene Expression Regulation, Enzymologic , Hepatocytes/drug effects , Hepatocytes/metabolism , Histamine Antagonists/administration & dosage , Histamine Antagonists/pharmacokinetics , Histamine Antagonists/pharmacology , Male , Membrane Glycoproteins , Receptors, Interleukin-1
6.
Nucl Med Biol ; 40(7): 912-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23906667

ABSTRACT

The fatty acids, n-butyric acid (BA), 4-phenylbutyric acid (PBA) and valproic acid (VPA, 2-propylpentanoic acid) have been used for many years in the treatment of a variety of CNS and peripheral organ diseases including cancer. New information that these drugs alter epigenetic processes through their inhibition of histone deacetylases (HDACs) has renewed interest in their biodistribution and pharmacokinetics and the relationship of these properties to their therapeutic and side effect profiles. In order to determine the pharmacokinetics and biodistribution of these drugs in primates, we synthesized their carbon-11 labeled analogues and performed dynamic positron emission tomography (PET) in six female baboons over 90 min. The carbon-11 labeled carboxylic acids were prepared by using (11)CO2 and the appropriate Grignard reagents. [(11)C]BA was metabolized rapidly (only 20% of the total carbon-11 in plasma was parent compound at 5 min post injection) whereas for VPA and PBA 98% and 85% of the radioactivity were the unmetabolized compound at 30 min after their administration respectively. The brain uptake of all three carboxylic acids was very low (<0.006%ID/cc, BA>VPA>PBA), which is consistent with the need for very high doses for therapeutic efficacy. Most of the radioactivity was excreted through the kidneys and accumulated in the bladder. However, the organ biodistribution between the drugs differed. [(11)C]BA showed relatively high uptake in spleen and pancreas whereas [(11)C]PBA showed high uptake in liver and heart. Notably, [(11)C]VPA showed exceptionally high heart uptake possibly due to its involvement in lipid metabolism. The unique biodistribution of each of these drugs may be of relevance in understanding their therapeutic and side effect profile including their teratogenic effects.


Subject(s)
Histone Deacetylase Inhibitors/pharmacokinetics , Positron-Emission Tomography , Animals , Blood Proteins/metabolism , Brain/diagnostic imaging , Brain/metabolism , Butyric Acid/blood , Butyric Acid/metabolism , Butyric Acid/pharmacokinetics , Carbon Radioisotopes , Female , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/metabolism , Isotope Labeling , Papio , Phenylbutyrates/blood , Phenylbutyrates/metabolism , Phenylbutyrates/pharmacokinetics , Radiochemistry , Tissue Distribution , Valproic Acid/blood , Valproic Acid/metabolism , Valproic Acid/pharmacokinetics
7.
Lipids ; 46(3): 277-85, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21225371

ABSTRACT

Butyric acid has been the subject of much attention last years due to its bioactivity. However, the potential advantages of butyrate are limited by the problem to reach enough plasma concentrations; therefore, pro-drugs have been proposed as an alternative to natural butyrate. A comparative study on in vitro intestinal digestion of 2,3-dibutyroil-1-O-octadecyl glycerol (D-SCAKG) and tributyrin (TB), as potential pro-drugs of butyric acid, was performed. Aliquots were taken at different times of digestion for studying the extent and rate of hydrolysis of both substrates. The micellar phase (MP) and oily phase (OP) formed in the digestion media were separated and their composition in lipid products was analyzed. Initially, it was confirmed that the in vitro model reproduced physiological results by testing against olive oil as a standard lipid. The progress of in vitro intestinal digestion of D-SCAKG was slower than that of TB. TB hydrolyzed completely to butyric acid, whereas D-SCAKG mainly yielded 2-butyroil-1-O-octadecyl glycerol (M-SCAKG), followed by butyric acid and 1-O-octadecyl glycerol (AKG). The MP from both substrates mainly consisted of butyric acid. Minor levels of M-SCAKG and AKG were also found in the MP after hydrolysis of D-SCAKG, the M-SCAKG being mainly distributed in the OP. Therefore, D-SCAKG produced a stable form of esterified butyric acid as M-SCAKG after in vitro intestinal digestion, unlike TB. Additionally, such a product would integrate both bioactive compounds, butyric acid and alkylglycerol, within the same molecule. Free butyric acid and AKG would be also released, which are lipid products of interest as well.


Subject(s)
Butyric Acid/administration & dosage , Glycerol/pharmacokinetics , Intestinal Mucosa/metabolism , Pharmaceutical Vehicles/pharmacokinetics , Triglycerides/pharmacokinetics , Biological Availability , Butyric Acid/chemistry , Butyric Acid/pharmacokinetics , Digestion/physiology , Diglycerides/chemistry , Diglycerides/pharmacokinetics , Drug Compounding , Glyceryl Ethers/chemistry , Glyceryl Ethers/pharmacokinetics , Humans , Hydrolysis , In Vitro Techniques , Models, Biological , Triglycerides/chemistry
8.
Toxicol Sci ; 108(1): 173-83, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18940962

ABSTRACT

Occupational exposure to butter flavoring vapors (BFV) is associated with significant pulmonary injury. The goal of the current study was to characterize inhalation dosimetric patterns of diacetyl and butyric acid, two components of BFV, and to develop a hybrid computational fluid dynamic-physiologically based pharmacokinetic model (CFD-PBPK) to describe these patterns. Uptake of diacetyl and butyric acid vapors, alone and in combination, was measured in the upper respiratory tract of anesthetized male Sprague-Dawley rats under constant velocity flow conditions and the uptake data were used to validate the CFD-PBPK model. Diacetyl vapor (100 or 300 ppm) was scrubbed from the airstream with 76-36% efficiency at flows of 100-400 ml/min. Butryic acid (30 ppm) was scrubbed with >90% efficiency. Concurrent exposure to butyric acid resulted in a small but significant reduction of diacetyl uptake (36 vs. 31%, p < 0.05). Diacetyl was metabolized in nasal tissues in vitro, likely by diacetyl reductase, an enzyme known to be inhibited by butyric acid. The CFD-PBPK model closely described diacetyl uptake; the reduction in diacetyl uptake by butyric acid could be explained by inhibition of diacetyl reductase. Extrapolation to the human via the model suggested that inspired diacetyl may penetrate to the intrapulmonary airways to a greater degree in the human than in the rat. Thus, based on dosimetric relationships, extrapulmonary airway injury in the rat may be predictive of intrapulmonary airway injury in humans. Butyric acid may modulate diacetyl toxicity by inhibiting its metabolism and/or altering its inhalation dosimetric patterns.


Subject(s)
Butyric Acid/pharmacokinetics , Computer Simulation , Diacetyl/pharmacokinetics , Inhalation Exposure/analysis , Models, Biological , Analysis of Variance , Animals , Biomarkers, Pharmacological , Butyric Acid/administration & dosage , Diacetyl/administration & dosage , Dose-Response Relationship, Drug , Humans , Male , NADP/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/metabolism , Sensitivity and Specificity
9.
Toxicol Sci ; 85(1): 429-46, 2005 May.
Article in English | MEDLINE | ID: mdl-15703268

ABSTRACT

The metabolic series approach for risk assessment uses a dosimetry-based analysis to develop toxicity information for a group of metabolically linked compounds using pharmacokinetic (PK) data for each compound and toxicity data for the parent compound. The metabolic series approach for n-butyl acetate and its subsequent metabolites, n-butanol and n-butyric acid (the butyl series), was first demonstrated using a provisional physiologically based pharmacokinetic (PBPK) model for the butyl series. The objective of this work was to complete development of the PBPK model for the butyl series. Rats were administered test compounds by iv bolus dose, iv infusion, or by inhalation in a recirculating closed chamber. Hepatic, vascular, and extravascular metabolic constants for metabolism were estimated by fitting the model to the blood time course data from these experiments. The respiratory bioavailability of n-butyl acetate (100% of alveolar ventilation) and n-butanol (50% of alveolar ventilation) was estimated from closed chamber inhalation studies and measured ventilation rates. The resulting butyl series PBPK model successfully reproduces the blood time course of these compounds following iv administration and inhalation exposure to n-butyl acetate and n-butanol in rats and arterial blood n-butanol kinetics following inhalation exposure to n-butanol in humans. These validated inhalation route models can be used to support species and dose-route extrapolations required for risk assessment of butyl series family of compounds. Human equivalent concentrations of 169 ppm and 1066 ppm n-butanol corresponding to the rat n-butyl acetate NOAELs of 500 and 3000 ppm were derived using the models.


Subject(s)
1-Butanol/pharmacokinetics , Acetates/pharmacokinetics , Butyric Acid/pharmacokinetics , Models, Biological , 1-Butanol/blood , Acetates/blood , Administration, Inhalation , Animals , Butyric Acid/blood , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Rats , Rats, Sprague-Dawley , Risk Assessment , Tissue Distribution
10.
Eur J Pharm Biopharm ; 58(3): 673-80, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15451544

ABSTRACT

Solid lipid nanoparticles (SLN) carrying cholesteryl butyrate (chol-but), doxorubicin and paclitaxel had previously been developed, and the antiproliferative effect of SLN formulations versus conventional drug formulations was here evaluated on HT-29 cells. The 50% inhibitory concentration (IC(50) values were interpolated from growth curves obtained by trypan blue exclusion assay. In vitro cytotoxicity of SLN carrying chol-but (IC(50 72 h) 0.3 +/- 0.03 mM vs >0.6 mM) and doxorubicin (IC(50 72 h) 81.87 +/- 4.11 vs 126.57 +/- 0.72 nM) was higher than that of conventional drug formulations. Intracellular doxorubicin was double after 24 h exposure to loaded SLN versus the conventional drug formulation, at the highest concentration evaluated by flow cytometry. In vitro cytotoxicities of paclitaxel-loaded SLN and conventional drug formulation (IC(50 72 h) 37.36 +/- 6.41 vs 33.43 +/-1.17 nM) were similar. Moreover, the combination of low concentrations of chol-but SLN (0.1-0.2 mM) and doxorubicin (1.72 nM) or paclitaxel (1.17 nM) exerted a greater-than-additive antiproliferative effect at 24 h exposure, while the combination of Na-but and doxorubicin or paclitaxel did not. These preliminary in vitro results suggest that SLN could be proposed as alternative drug delivery system.


Subject(s)
Antineoplastic Agents/toxicity , Nanostructures/toxicity , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Butyric Acid/administration & dosage , Butyric Acid/pharmacokinetics , Butyric Acid/toxicity , Cell Survival/drug effects , Cell Survival/physiology , Cholesterol Esters/administration & dosage , Cholesterol Esters/pharmacokinetics , Cholesterol Esters/toxicity , Colorectal Neoplasms/drug therapy , Dose-Response Relationship, Drug , HT29 Cells , Humans
11.
Clin Cancer Res ; 10(14): 4822-30, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15269158

ABSTRACT

PURPOSE: The purpose is to evaluate the CD44-mediated cellular targeting of HA-But, a hyaluronic acid esterified with butyric acid (But) residues, to hepatocellular carcinoma cell lines in vitro and to hepatic tumor metastases in vivo. EXPERIMENTAL DESIGN: In vitro, the CD44-dependent cytotoxicity in two human hepatocellular carcinoma cell lines (HepB3 and HepG2) with high and low CD44 expression was investigated; in vivo, the effect on liver metastases originating from intrasplenic implants of Lewis lung carcinoma (LL3) or B16-F10 melanoma in mice was compared with the pharmacokinetics of organ and tissue distribution using different routes of administration. RESULTS: HepB3 and HepG2 cell lines showed different expression of CD44 (78 and 18%, respectively), which resulted in a CD44-dependent HA-But inhibitory effect as demonstrated also by the uptake analysis performed using radiolabeled HA-But ((99m)Tc-HA-But). Pharmacokinetic studies showed different rates of (99m)Tc-HA-But distribution according to the route of administration (i.v., i.p., or s.c.): very fast (a few minutes) after i.v. treatment, with substantial accumulation in the liver and spleen; relatively slow after i.p. or s.c. treatment, with marked persistence of the drug at the site of injection. The effect of s.c. and i.p. treatment with HA-But on liver metastases originating from intrasplenic implants of LL3 carcinoma or B16-F10 melanoma (both CD44-positive: 68 and 87%, respectively), resulted in 87 and 100% metastases-free animals, respectively (regardless of the route of administration), and a significant prolongation of the life expectancy compared with control groups. CONCLUSIONS: HA-But tends to concentrate in the liver and spleen and appears to be a promising new drug for the treatment of intrahepatic tumor lesions.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Liver Neoplasms/prevention & control , Animals , Butyric Acid/chemistry , Butyric Acid/pharmacokinetics , Butyric Acid/pharmacology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/prevention & control , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Esters , Female , Flow Cytometry , Humans , Hyaluronan Receptors/analysis , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacokinetics , Hyaluronic Acid/pharmacology , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/pharmacokinetics , Survival Analysis , Tissue Distribution
12.
Am J Physiol Gastrointest Liver Physiol ; 286(2): G197-203, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14525727

ABSTRACT

Butyrate and the other short-chain fatty acids (SCFAs) are the most abundant anions in the colonic lumen. Also, butyrate is the preferred energy source for colonocytes and has been shown to regulate colonic electrolyte and fluid absorption. Previous studies from our group have demonstrated that the HCO(3)(-)/SCFA(-) anion exchange process is one of the major mechanisms of butyrate transport across the purified human colonic apical membrane vesicles and the apical membrane of human colonic adenocarcinoma cell line Caco-2 and have suggested that it is mainly mediated via monocarboxylate transporter-1 (MCT-1) isoform. However, little is known regarding the regulation of SCFA transport by various hormones and signal transduction pathways. Therefore, the present studies were undertaken to examine whether hydrocortisone and phorbol 12-myristate 13-acetate (PMA) are involved in a possible regulation of the butyrate/anion exchange process in Caco-2 cells. The butyrate/anion exchange process was assessed by measuring a pH-driven [(14)C]butyrate uptake in Caco-2 cells. Our results demonstrated that 24-h incubation with PMA (1 microM) significantly increased [(14)C]butyrate uptake compared with incubation with 4alphaPMA (inactive form). In contrast, incubation with hydrocortisone had no significant effect on butyrate uptake in Caco-2 cells compared with vehicle (ethanol) alone. Induction of butyrate uptake by PMA appeared to be via an increase in the maximum velocity (V(max)) of the transport process with no significant changes in the K(m) of the transporter for butyrate. Parallel to the increase in the V(max) of [(14)C]butyrate uptake, the MCT-1 protein level was also increased in response to PMA incubation. Our studies demonstrated that the butyrate/anion exchange was increased in response to PMA treatment along with the induction in the level of MCT-1 expression in Caco-2 cells.


Subject(s)
Adenocarcinoma/metabolism , Butyric Acid/pharmacokinetics , Carcinogens/pharmacology , Colonic Neoplasms/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Adenocarcinoma/pathology , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Colonic Neoplasms/pathology , Drug Administration Schedule , Humans , Hydrocortisone/pharmacology , Lactic Acid/pharmacology , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/physiology , Symporters/metabolism , Symporters/physiology , Tetradecanoylphorbol Acetate/administration & dosage
13.
J Nutr ; 133(10): 3145-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14519799

ABSTRACT

In humans, colonic bacteria ferment unabsorbed carbohydrates, producing the SCFA acetic, propionic and n-butyric acids. To test for interactions among the SCFA that may affect their absorption, healthy subjects (n = 10) were given 300-mL rectal infusions containing acetate (60 mmol/L), propionate (20 mmol/L) and butyrate (20 mmol/L), alone or in combinations of two or three. The solutions were retained for 30 min, and then subjects voided a sample for SCFA measurement. To examine the relationship between absorption and fecal SCFA concentrations, a fecal sample was collected at the end of the study. The mean percentage of butyrate absorption (30.2 +/- 4.6%) exceeded that of acetate (24.1 +/- 3.7%) (P < 0.05). Absorption tended to be less (P = 0.12) when a SCFA was infused alone (26.7 +/- 4.0%) than when all three were infused (32.0 +/- 5.7%). Bicarbonate concentration was higher after butyrate-containing infusions than after saline. The fecal molar acetate percentage was inversely correlated with the percentage of acetate absorption from the infusion of three SCFA (r = -0.834, P < 0.005). We conclude that there was no combination effect on SCFA absorption, and the chain-length effect suggests passive diffusion as a likely mechanism of absorption. Furthermore, fecal acetate may reflect absorption, rather than production of colonic acetate.


Subject(s)
Acetic Acid/analysis , Acetic Acid/pharmacokinetics , Colon/metabolism , Feces/chemistry , Intestinal Absorption , Rectum/metabolism , Acetic Acid/administration & dosage , Adult , Bicarbonates/analysis , Breath Tests , Butyric Acid/administration & dosage , Butyric Acid/analysis , Butyric Acid/pharmacokinetics , Female , Humans , Male , Methane/analysis , Propionates/administration & dosage , Propionates/analysis , Propionates/pharmacokinetics , Solutions
14.
Methods Find Exp Clin Pharmacol ; 23(5): 245-53, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11712644

ABSTRACT

Few pharmaceutical studies, with the exception of those on rectal solutions, are described on short chain fatty acid (SCFA) formulations-especially for sodium butyrate, which is a colonocyte preferential substrate. Highly dosed butyrate pellets (90%) were prepared and their coating was designed for colonic delivery. In vivo determination (pH and transit time of pellets in rats) allowed to respectively choose the grade and thickness (resistance of 6 h) of the pH-dependent coating (Eudragi L+S, 1:1). The coated pellets were administered to naturally butyrate-deprived rats. The rats' colonic mucosa had the particularity to weakly express mitochondrial HMG-CoA synthase, an enzyme that responds to luminal butyrate. The results did not show early absorption of butyrate, but a probable cecal loss in the rat cecum as cecal residence time of the pellets was important and as pH was propitious for the coating hydrolysis. It seemed that butyrate, given daily for 7 days without the other main SCFA. was unable to induce the enzyme and/or that the dose (0.32 mmol/day) was insufficient.


Subject(s)
Butyric Acid/administration & dosage , Colon/metabolism , Polymethacrylic Acids , Administration, Oral , Animals , Butyric Acid/pharmacokinetics , Carmine/pharmacokinetics , Cecum/chemistry , Coenzyme A Ligases/metabolism , Colon/cytology , Colon/enzymology , Coloring Agents , Gastric Mucosa/chemistry , Gastric Mucosa/enzymology , Gastrointestinal Contents/chemistry , Gastrointestinal Transit , Hydrogen-Ion Concentration , Hydroxymethylglutaryl-CoA Synthase , In Vitro Techniques , Norepinephrine/pharmacokinetics , Rats , Rats, Inbred F344 , Tablets
15.
J Pharm Sci ; 89(11): 1486-95, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11015693

ABSTRACT

6(A)-O-(n-Butanoyl)-beta-cyclodextrin was prepared and its hydrolysis behavior in aqueous solutions and in rat intestinal fluids was investigated. Furthermore, the enzymatic hydrolyses of the n-butyric acid-beta-cyclodextrin conjugate using alpha-amylase and esterase were studied to gain insight into the release behavior of n-butyric acid from the conjugate. The hydrolysis of the conjugate proceeded according to a first-order kinetics in aqueous solution, and gave a V-shaped pH profile, indicating a specific acid-base-catalyzed hydrolysis at acidic and neutral-alkaline regions, respectively. The half-lives (t(1/2)) of the conjugate at pH 4.4, 6.8, and 7.4 at 37 degrees C were approximately 580, 43, and 6 days, respectively, indicating that the conjugate is stable in aqueous solution. No appreciable release of n-butyric acid from the conjugate was observed in the stomach and small intestinal contents of rats, or in the small and large intestinal homogenates of rats. On the other hand, a fast disappearance of the conjugate and an appearance of n-butyric acid were observed in the cecal and colonic contents of rats. The t(1/2) values of the disappearance were approximately 4, 1, and 6 h in 10 and 15% cecal contents and 10% colonic contents, respectively, and the appearance of n-butyric acid after 6 h was approximately 10% in the 15% cecal contents. Aspergillus oryzae alpha-amylase hydrolyzed the conjugate to small saccharide conjugates, such as the triose and maltose conjugates, but there was no appreciable release of n-butyric acid. The conjugate was less susceptible to carboxylic esterase (from porcine live), thus releasing no appreciable amounts of n-butyric acid. On the other hand, a fast release of n-butyric acid was observed when the esterase was employed after amylase hydrolysis, suggesting that two types of enzymes, sugar-degrading and ester-hydrolyzing enzymes, are necessary for the release of n-butyric acid from the conjugate in large intestinal contents.


Subject(s)
Butyric Acid/pharmacokinetics , Cyclodextrins/pharmacokinetics , Food Additives/pharmacokinetics , Histamine Antagonists/pharmacokinetics , Intestinal Mucosa/metabolism , beta-Cyclodextrins , Animals , Butyric Acid/chemistry , Culture Media , Cyclodextrins/chemistry , Esters/chemistry , Esters/pharmacokinetics , Fatty Acids/chemistry , Fatty Acids/pharmacokinetics , Food Additives/chemistry , Histamine Antagonists/chemistry , Male , Rats , Rats, Wistar , Solubility
16.
Anticancer Res ; 19(5B): 3921-5, 1999.
Article in English | MEDLINE | ID: mdl-10628332

ABSTRACT

BACKGROUND: Cholesteryl-butyrate chosen as lipid matrix of solid lipid nanospheres (SLNs) could be a suitable pro-drug to deliver butyric acid and overcome one of the most limiting disadvantages of the compound: the short half-life due to a rapid metabolism. METHODS: We evaluated the antiproliferative effect, with respect to that of sodium butyrate, of four SLNs (SLN1, SLN2, SLN3 and SLN4) characterized by a different concentration of cholesteryl-butyrate (range, 1.7-30 mM) on NIH-H460, a non-small cell lung carcinoma cell line. RESULTS: After 6 days of treatment, all SLN preparations induced a dose-dependent inhibition of NHI-H460 cell growth: the most effective SLN preparation (SLN1) was able to induce a complete growth inhibition already at 0.25 mM, a concentration at which sodium butyrate induced only a 55% inhibition. Fluorescence microscopy showed that 6-coumarin-tagged SLNs were almost completely internalized by cells after 5 min of treatment. CONCLUSIONS: The present results indicate that owing to their peculiar characteristics, SLNs could be an interesting alternative approach for butyric acid delivery into tumor cells.


Subject(s)
Butyrates/pharmacokinetics , Butyric Acid/pharmacokinetics , Cholesterol/pharmacokinetics , Drug Carriers , Drug Carriers/pharmacokinetics , Lipids/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Humans , Lipids/chemistry , Lung Neoplasms/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Phosphatidylcholines/pharmacokinetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL