Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 163: 114860, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37196540

ABSTRACT

The antibiotic-induced intestinal injury (AIJ) is associated with diarrhoea and gastrointestinal discomfort. However, the pathological intestinal mechanisms and related side effects associated with antibiotic use/misuse may be counteracted by probiotics. This study aims to evaluate the effect and the protective mechanisms of a probiotic formulation containing Alkalihalobacillus clausii (formerly Bacillus clausii; BC) spores in an experimental model of AIJ. C57/Bl6J mice were orally challenged with a high dose of ceftriaxone for five days along with BC treatment which lasted up to the 15th day. Our results showed the beneficial effect of the probiotic in preserving colonic integrity and limiting tissue inflammation and immune cell infiltration in AIJ mice. BC increased tight junction expression and regulated the unbalanced production of colonic pro- and anti-inflammatory cytokines, converging toward the full resolution of the intestinal damage. These findings were supported by the histological evaluation of the intestinal mucosa, suggesting a potential restoration of mucus production. Notably, BC treatment increased gene transcription of the secretory products responsible for epithelium repair and mucus synthesis and normalized the expression of antimicrobial peptides involved in immune activation. Reconstruction of complex and diverse gut microbiota in antibiotic-induced dysbiosis was recorded upon BC supplementation. Specifically, the expansion of A. clausii, Prevotella rara and Eubacterium ruminatium drove intestinal microbiota rebalance by primarily impacting Bacteroidota members. Taken together, our data indicate that BC administration alleviates AIJ by multiple converging mechanisms leading to restoring gut integrity and homeostasis and reshaping microbiota composition.


Subject(s)
Bacillus clausii , Gastrointestinal Microbiome , Intestinal Diseases , Probiotics , Animals , Mice , Anti-Bacterial Agents/therapeutic use , Bacillus clausii/physiology , Spores, Bacterial , Intestinal Diseases/drug therapy , Intestinal Mucosa , Probiotics/pharmacology
2.
Biomed Pharmacother ; 153: 113528, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36076609

ABSTRACT

Paclitaxel (PTX) is one of the most broadly used chemotherapeutic agents for the treatment of several tumor types including ovarian, breast, and non-small cell lung cancer. However, its use is limited by debilitating side effects, involving both gastrointestinal and behavioral dysfunctions. Due to growing evidence showing a link between impaired gut function and chemotherapy-associated behavioral changes, the aim of this study was to identify a novel therapeutic approach to manage PTX-induced gut and brain comorbidities. Mice were pre-treated with sodium butyrate (BuNa) for 30 days before receiving PTX. After 14 days, mice underwent to behavioral analysis and biochemical investigations of gut barrier integrity and microbiota composition. Paired evaluations of gut functions revealed that the treatment with BuNa restored PTX-induced altered gut barrier integrity, microbiota composition and food intake suggesting a gut-to-brain communication. The treatment with BuNa also ameliorated depressive- and anxiety-like behaviors induced by PTX in mice, and these effects were associated with neuroprotective and anti-inflammatory outcomes. These results propose that diet supplementation with this safe postbiotic might be considered when managing PTX-induced central side effects during cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Microbiome , Intestinal Diseases , Lung Neoplasms , Animals , Butyric Acid/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Dietary Supplements , Intestinal Diseases/drug therapy , Lung Neoplasms/drug therapy , Mice , Mice, Inbred C57BL , Paclitaxel/adverse effects
3.
Br J Pharmacol ; 174(12): 1704-1718, 2017 06.
Article in English | MEDLINE | ID: mdl-26758851

ABSTRACT

The intestine is composed of many distinct cell types that respond to commensal microbiota or pathogens with immune tolerance and proinflammatory signals respectively. ROS produced by mucosa-resident cells or by newly recruited innate immune cells are essential for antimicrobial responses and regulation of signalling pathways including processes involved in wound healing. Impaired ROS production due to inactivating patient variants in genes encoding NADPH oxidases as ROS source has been associated with Crohn's disease and pancolitis, whereas overproduction of ROS due to up-regulation of oxidases or altered mitochondrial function was linked to ileitis and ulcerative colitis. Here, we discuss recent advances in our understanding of how maintaining a redox balance is crucial to preserve gut homeostasis. LINKED ARTICLES: This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.


Subject(s)
Gastrointestinal Tract/metabolism , Inflammation/metabolism , Reactive Oxygen Species/metabolism , Animals , Gastrointestinal Tract/pathology , Homeostasis , Humans , Inflammation/pathology , Oxidation-Reduction
4.
Mucosal Immunol ; 7(1): 57-67, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23612054

ABSTRACT

MyD88 adapter-like (Mal)-deficient mice displayed increased susceptibility to oral but not intraperitoneal infection with Salmonella Typhimurium. Bone marrow chimeras demonstrated that mice with Mal-deficient non-hematopoietic cells were more susceptible to infection, indicating a role for Mal in non-myeloid cells. We observed perturbed barrier function in Mal(-/-) mice, as indicated by reduced electrical resistance and increased mucosa blood permeability following infection. Altered expression of occludin, Zonula occludens-1, and claudin-3 in intestinal epithelia from Mal(-/-) mice suggest that Mal regulates tight junction formation, which may in part contribute to intestinal integrity. Mal interacted with several protein kinase C (PKC) isoforms in a Caco-2 model of intestinal epithelia and inhibition of Mal or PKC increased permeability and bacterial invasion via a paracellular route, while a pan-PKC inhibitor increased susceptibility to oral infection in mice. Mal signaling is therefore beneficial to the integrity of the intestinal barrier during infection.


Subject(s)
Intestinal Mucosa/metabolism , Membrane Glycoproteins/metabolism , Protein Kinase C/metabolism , Receptors, Interleukin-1/metabolism , Animals , Cell Line , Gene Expression Regulation , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Permeability , Protein Binding , Protein Transport , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Signal Transduction , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
5.
Acta Physiol (Oxf) ; 204(2): 255-66, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21726418

ABSTRACT

AIM: Plant cannabinoids, like Δ(9)-tetrahydrocannabinol (THC) and cannabidiol (CBD), activate/desensitize thermosensitive transient receptor potential (TRP) channels of vanilloid type-1 or -2 (TRPV1 or TRPV2). We investigated whether cannabinoids also activate/desensitize two other 'thermo-TRP's', the TRP channels of vanilloid type-3 or -4 (TRPV3 or TRPV4), and if the TRPV-inactive cannabichromene (CBC) modifies the expression of TRPV1-4 channels in the gastrointestinal tract. METHODS: TRP activity was assessed by evaluating elevation of [Ca(2+)](i) in rat recombinant TRPV3- and TRPV4-expressing HEK-293 cells. TRP channel mRNA expression was measured by quantitative RT-PCR in the jejunum and ileum of mice treated with vehicle or the pro-inflammatory agent croton oil. RESULTS: (i) CBD and tetrahydrocannabivarin (THCV) stimulated TRPV3-mediated [Ca(2+)](i) with high efficacy (50-70% of the effect of ionomycin) and potency (EC(50∼) 3.7 µm), whereas cannabigerovarin (CBGV) and cannabigerolic acid (CBGA) were significantly more efficacious at desensitizing this channel to the action of carvacrol than at activating it; (ii) cannabidivarin and THCV stimulated TRPV4-mediated [Ca(2+)](i) with moderate-high efficacy (30-60% of the effect of ionomycin) and potency (EC(50) 0.9-6.4 µm), whereas CBGA, CBGV, cannabinol and cannabigerol were significantly more efficacious at desensitizing this channel to the action of 4-α-phorbol 12,13-didecanoate (4α-PDD) than at activating it; (iii) CBC reduced TRPV1ß, TRPV3 and TRPV4 mRNA in the jejunum, and TRPV3 and TRPV4 mRNA in the ileum of croton oil-treated mice. CONCLUSIONS: Cannabinoids can affect both the activity and the expression of TRPV1-4 channels, with various potential therapeutic applications, including in the gastrointestinal tract.


Subject(s)
Cannabinoids/pharmacology , Gastrointestinal Diseases/metabolism , Intestine, Small/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cannabidiol/chemistry , Cannabidiol/pharmacology , Cannabinoids/chemistry , Dronabinol/chemistry , Dronabinol/pharmacology , Gastrointestinal Diseases/chemically induced , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Mice , Rats , TRPV Cation Channels/genetics
6.
Neurogastroenterol Motil ; 23(8): 745-e331, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21689210

ABSTRACT

BACKGROUND: Bromelain (BR) is a cysteine protease with inhibitory effects on intestinal secretion and inflammation. However, its effects on intestinal motility are largely unexplored. Thus, we investigated the effect of this plant-derived compound on intestinal contractility and transit in mice. METHODS: Contractility in vitro was evaluated by stimulating the mouse isolated ileum, in an organ bath, with acetylcholine, barium chloride, or electrical field stimulation. Motility in vivo was measured by evaluating the distribution of an orally administered fluorescent marker along the small intestine. Transit was also evaluated in pathophysiologic states induced by the pro-inflammatory compound croton oil or by the diabetogenic agent streptozotocin. KEY RESULTS: Bromelain inhibited the contractions induced by different spasmogenic compounds in the mouse ileum with similar potency. The antispasmodic effect was reduced or counteracted by the proteolytic enzyme inhibitor, gabexate (15 × 10(-6) mol L(-1) ), protease-activated receptor-2 (PAR-2) antagonist, N(1) -3-methylbutyryl-N(4) -6-aminohexanoyl-piperazine (10(-4) mol L(-1) ), phospholipase C (PLC) inhibitor, neomycin (3 × 10(-3) mol L(-1) ), and phosphodiesterase 4 (PDE4) inhibitor, rolipram (10(-6) mol L(-1) ). In vivo, BR preferentially inhibited motility in pathophysiologic states in a PAR-2-antagonist-sensitive manner. CONCLUSIONS & INFERENCES: Our data suggest that BR inhibits intestinal motility - preferentially in pathophysiologic conditions - with a mechanism possibly involving membrane PAR-2 and PLC and PDE4 as intracellular signals. Bromelain could be a lead compound for the development of new drugs, able to normalize the intestinal motility in inflammation and diabetes.


Subject(s)
Ananas/enzymology , Bromelains/pharmacology , Gastrointestinal Motility/drug effects , Gastrointestinal Transit/drug effects , Acetylcholine/pharmacology , Animals , Barium Compounds/pharmacology , Bromelains/metabolism , Caco-2 Cells , Chlorides/pharmacology , Cholinergic Agonists/pharmacology , Croton Oil/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Electric Stimulation , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Ileitis/chemically induced , Ileitis/physiopathology , Male , Mice , Muscle Contraction/drug effects , Peptides/metabolism , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-2/antagonists & inhibitors
7.
Eur Rev Med Pharmacol Sci ; 12 Suppl 1: 81-93, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18924447

ABSTRACT

The plant Cannabis has been known for centuries to be beneficial in a variety of gastrointestinal diseases, including emesis, diarrhea, inflammatory bowel disease and intestinal pain. delta9-tetrahydrocannabinol, the main psychotropic component of Cannabis, acts via at least two types of cannabinoid receptors, named CB1 and CB2 receptors. CB1 receptors are located primarily on central and peripheral neurons (including the enteric nervous system) where they modulate neurotransmitter release, whereas CB2 receptors are concerned with immune function, inflammation and pain. The discovery of endogenous ligands [i.e. anandamide and 2-arachidonoyl glycerol (2-AG)] for these receptors indicates the presence of a functional endogenous cannabinoid system in the gastrointestinal tract. Anatomical and functional evidence suggests the presence of CB1 receptors in the myenteric plexus, which are associated with cholinergic neurons in a variety of species, including in humans. Activation of prejunctional CB1 receptors reduces excitatory enteric transmission (mainly cholinergic transmission) in different regions of the gastrointestinal tract. Consistently, in vivo studies have shown that cannabinoids reduce gastrointestinal transit in rodents through activation of CB1, but not CB2, receptors. However, in pathophysiological states, both CB1 and CB2 receptors could reduce the increase of intestinal motility induced by inflammatory stimuli. Cannabinoids also reduce gastrointestinal motility in randomized clinical trials. Overall, modulation of the gut endogenous cannabinoid system may provide a useful therapeutic target for disorders of gastrointestinal motility.


Subject(s)
Cannabinoids/pharmacology , Gastrointestinal Motility/drug effects , Receptors, Cannabinoid/drug effects , Animals , Cannabis/chemistry , Gastrointestinal Diseases/drug therapy , Humans , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/physiopathology , Synaptic Transmission/drug effects
8.
Br J Pharmacol ; 155(5): 681-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18622408

ABSTRACT

BACKGROUND AND PURPOSE: Salvinorin A, the active component of the hallucinogenic herb Salvia divinorum, inhibits intestinal motility through activation of kappa-opioid receptors (KORs). However, this compound may have target(s) other than the KORs in the inflamed gut. Because intestinal inflammation upregulates cannabinoid receptors and endogenous cannabinoids, in the present study we investigated the possible involvement of the endogenous cannabinoid system in salvinorin A-induced delay in motility in the inflamed gut. EXPERIMENTAL APPROACH: Motility in vivo was measured by evaluating the distribution of a fluorescent marker along the small intestine; intestinal inflammation was induced by the irritant croton oil; direct or indirect activity at cannabinoid receptors was evaluated by means of binding, enzymic and cellular uptake assays. KEY RESULTS: Salvinorin A as well as the KOR agonist U-50488 reduced motility in croton oil treated mice. The inhibitory effect of both salvinorin A and U-50488 was counteracted by the KOR antagonist nor-binaltorphimine and by the cannabinoid CB(1) receptor antagonist rimonabant. Rimonabant, however, did not counteract the inhibitory effect of salvinorin A on motility in control mice. Binding experiments showed very weak affinity of salvinorin A for cannabinoid CB(1) and CB(2) and no inhibitory effect on 2-arachidonoylglycerol and anandamide hydrolysis and cellular uptake. CONCLUSIONS AND IMPLICATIONS: The inhibitory effect of salvinorin A on motility reveals a functional interaction between cannabinoid CB(1) receptors and KORs in the inflamed--but not in the normal--gut in vivo.


Subject(s)
Diterpenes, Clerodane/pharmacology , Gastrointestinal Motility/drug effects , Ileitis/physiopathology , Receptor Cross-Talk/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, kappa/metabolism , Salvia/chemistry , Amidohydrolases/biosynthesis , Animals , COS Cells , Chlorocebus aethiops , Croton Oil/pharmacology , Diterpenes, Clerodane/isolation & purification , Electric Stimulation , Ileitis/chemically induced , Ileitis/enzymology , Ileitis/metabolism , Intestine, Small/drug effects , Intestine, Small/enzymology , Intestine, Small/metabolism , Male , Mice , Mice, Inbred ICR , Plant Leaves/chemistry , Protein Binding , Receptor, Cannabinoid, CB2/metabolism , Receptors, Opioid, kappa/agonists
9.
Br J Pharmacol ; 154(5): 1001-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18469842

ABSTRACT

BACKGROUND AND PURPOSE: Cannabidiol is a Cannabis-derived non-psychotropic compound that exerts a plethora of pharmacological actions, including anti-inflammatory, neuroprotective and antitumour effects, with potential therapeutic interest. However, the actions of cannabidiol in the digestive tract are largely unexplored. In the present study, we investigated the effect of cannabidiol on intestinal motility in normal (control) mice and in mice with intestinal inflammation. EXPERIMENTAL APPROACH: Motility in vivo was measured by evaluating the distribution of an orally administered fluorescent marker along the small intestine; intestinal inflammation was induced by the irritant croton oil; contractility in vitro was evaluated by stimulating the isolated ileum, in an organ bath, with ACh. KEY RESULTS: In vivo, cannabidiol did not affect motility in control mice, but normalized croton oil-induced hypermotility. The inhibitory effect of cannabidiol was counteracted by the cannabinoid CB1 receptor antagonist rimonabant, but not by the cannabinoid CB2 receptor antagonist SR144528 (N-[-1S-endo-1,3,3-trimethyl bicyclo [2.2.1] heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide), by the opioid receptor antagonist naloxone or by the alpha2-adrenergic antagonist yohimbine. Cannabidiol did not reduce motility in animals treated with the fatty acid amide hydrolase (FAAH) inhibitor N-arachidonoyl-5-hydroxytryptamine, whereas loperamide was still effective. In vitro, cannabidiol inhibited ACh-induced contractions in the isolated ileum from both control and croton oil-treated mice. CONCLUSIONS AND IMPLICATIONS: Cannabidiol selectively reduces croton oil-induced hypermotility in mice in vivo and this effect involves cannabinoid CB1 receptors and FAAH. In view of its low toxicity in humans, cannabidiol may represent a good candidate to normalize motility in patients with inflammatory bowel disease.


Subject(s)
Cannabidiol/pharmacology , Cannabis , Gastrointestinal Agents/pharmacology , Gastrointestinal Motility/drug effects , Ileitis/drug therapy , Ileum/drug effects , Acetylcholine/pharmacology , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Cannabis/chemistry , Cholinergic Agents/pharmacology , Croton Oil , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gastrointestinal Transit/drug effects , Ileitis/chemically induced , Ileitis/physiopathology , Ileum/metabolism , Ileum/physiopathology , Loperamide/pharmacology , Male , Mice , Mice, Inbred ICR , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Rimonabant
10.
Br J Pharmacol ; 153(6): 1272-80, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18223666

ABSTRACT

BACKGROUND AND PURPOSE: Endocannabinoids (via cannabinoid CB(1) receptor activation) are physiological regulators of intestinal motility and food intake. However, their role in the regulation of gastric emptying is largely unexplored. The purpose of the present study was to investigate the involvement of the endocannabinoid system in the regulation of gastric emptying in mice fed either a standard diet (STD) or a high-fat diet (HFD) for 14 weeks. EXPERIMENTAL APPROACH: Gastric emptying was evaluated by measuring the amount of phenol red recovered in the stomach after oral challenge; CB(1) expression was analysed by quantitative reverse transcription-PCR; endocannabinoid (anandamide and 2-arachidonoyl glycerol) levels were measured by liquid chromatography-mass spectrometry. KEY RESULTS: Gastric emptying was reduced by anandamide, an effect counteracted by the CB(1) receptor antagonist rimonabant, but not by the CB(2) receptor antagonist SR144528 or by the transient receptor potential vanilloid type 1 (TRPV1) antagonist 5'-iodoresiniferatoxin. The fatty acid amide hydrolase (FAAH) inhibitor N-arachidonoyl-5-hydroxytryptamine (but not the anandamide uptake inhibitor OMDM-2) reduced gastric emptying in a way partly reduced by rimonabant. Compared to STD mice, HFD mice exhibited significantly higher body weight and fasting glucose levels, delayed gastric emptying and lower anandamide and CB(1) mRNA levels. N-arachidonoylserotonin (but not rimonabant) affected gastric emptying more efficaciously in HFD than STD mice. CONCLUSIONS AND IMPLICATIONS: Gastric emptying is physiologically regulated by the endocannabinoid system, which is downregulated following a HFD leading to overweight.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Dietary Fats/pharmacology , Down-Regulation , Endocannabinoids , Gastric Emptying/physiology , Animals , Arachidonic Acids/pharmacology , Blood Glucose , Body Weight , Chromatography, Liquid , Male , Mass Spectrometry , Mice , Mice, Inbred ICR , Phenolsulfonphthalein , Polyunsaturated Alkamides/pharmacology , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Neurogastroenterol Motil ; 20(2): 142-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17931335

ABSTRACT

The hallucinogenic plant Salvia divinorum has been used for medical treatments of gastrointestinal disorders. Here, we evaluated the effect of a standardized extract from the leaves of Salvia divinorum (SDE) and of its active ingredient salvinorin A on motility in vivo, both in physiological states and during croton oil-induced intestinal inflammation. SDE (1-100 mg kg(-1)) significantly inhibited motility only in inflamed, but not in control, mice. In control mice, salvinorin A (0.01-10 mg kg(-1)) significantly inhibited motility only at the highest doses tested (3 and 10 mg kg(-1)) and this effect was not counteracted by naloxone or by the kappa-opioid receptor (KOR) antagonist nor-binaltorphimine. Inflammation significantly increased the potency of salvinorin A (but not of the KOR agonist U-50488) in reducing motility. The inhibitory effects of both salvinorin A and U-50488 in inflamed mice were counteracted by naloxone or by nor-binaltorphimine. We conclude that salvinorin A may reduce motility through activation of different targets. In physiological states, salvinorin A, at high doses, inhibited motility through a non-KOR mediated mechanism. Gut inflammation increased the potency of salvinorin A; this effect was mediated by KOR, but it was not shared by U-50488, thus suggesting that salvinorin A may have target(s) other than KOR in the inflamed gut.


Subject(s)
Diterpenes/pharmacology , Gastrointestinal Motility/drug effects , Inflammation/physiopathology , Plant Extracts/pharmacology , Salvia/chemistry , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Diterpenes, Clerodane , Hallucinogens/pharmacology , Male , Mice , Mice, Inbred ICR , Plant Leaves/chemistry , Receptors, Opioid, kappa/drug effects
13.
Phytomedicine ; 12(6-7): 501-5; discussion 505, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16008128

ABSTRACT

Chronic use of anthraquinone laxatives has been blamed for the induction of habituation and the development of colonic cancer, but there are no definitive studies which have demonstrated this. To evaluate the carcinogenic potential of anthraquinones, the effect of long-term senna pod extract (SE) treatment on either healthy rats or rats treated with an initiating tumor agent (azoxymethane--AOM) has been studied. SE (30 and 60mg/kg), administered for 110 weeks, did not induce the development of aberrant crypt foci (ACF) and tumors in healthy rats. The development of ACF and tumors in rats treated with AOM were significantly reduced by SE (30 and 60 mg/kg). These results suggest that a chronic SE use does not predispose to colon cancer. By contrast, SE might exert an anti-tumoral activity on rat colon carcinogenesis.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/pharmacology , Colonic Neoplasms/prevention & control , Phytotherapy , Senna Extract/pharmacology , Senna Plant , Adenocarcinoma/chemically induced , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/therapeutic use , Azoxymethane , Cathartics , Colonic Neoplasms/chemically induced , Male , Precancerous Conditions/chemically induced , Precancerous Conditions/prevention & control , Rats , Rats, Wistar , Senna Extract/administration & dosage , Senna Extract/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...