Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Lett ; 15(5): 6688-6694, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29616131

RESUMO

The chemopreventive activity of non-steroidal anti-inflammatory drugs (NSAIDs), particularly aspirin, has been well demonstrated in preclinical and clinical studies. However, the primary side effect from this class of drug is gastrointestinal (GI) bleeding, which has limited the widespread use of NSAIDs for the prevention of cancer. The development of GI-safer NSAIDs, which are associated with phosphatidylcholine (PC) may provide a solution to this therapeutic problem. In the present study, the efficacy of two NSAIDs, aspirin and indomethacin, were compared using murine colon cancer cell line MC-26. Each NSAID was assessed alone and in combination with PC, using in vitro and in vivo systems. The results reveal that the PC-associated NSAIDs had a significantly higher degree of protection against cancer cell growth compared with the unmodified NSAIDs. It was also observed that Aspirin-PC and Indomethacin-PC prevented the metastatic spread of cancer cells in a syngeneic mouse model. These results support the potential use of PC-NSAIDs for the chemoprevention of colorectal cancer.

2.
Cancer Prev Res (Phila) ; 10(2): 142-152, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27998883

RESUMO

The mechanism by which aspirin consumption is linked to significant reductions in the incidence of multiple forms of cancer and metastatic spread to distant tissues, resulting in increased cancer patient survival is not well understood. In this study, using colon cancer as an example, we provide both in vitro (cell culture) and in vivo (chemically induced mouse model of colon cancer) evidence that this profound antineoplastic action may be associated with aspirin's ability to irreversibly inhibit COX-1-mediated platelet activation, thereby blocking platelet-cancer cell interactions, which promote cancer cell number and invasive potential. This process may be driven by platelet-induced epithelial-mesenchymal transition (EMT), as assessed using confocal microscopy, based upon changes in cell morphology, growth characteristics and fibronectin expression, and biochemical/molecular analysis by measuring changes in the expression of the EMT markers; vimentin, ß-catenin, and SNAIL. We also provide evidence that a novel, gastrointestinal-safe phosphatidylcholine (PC)-associated aspirin, PL2200 Aspirin, possesses the same or more pronounced actions versus unmodified aspirin with regard to antiplatelet effects (in vitro: reducing platelet activation as determined by measuring the release of thromboxane and VEGF in culture medium; in vivo: inhibiting platelet number/activation and extravasation into tumor tissue) and chemoprevention (in vitro: inhibiting colonic cell growth and invasive activity; in vivo: inhibiting colonic dysplasia, inflammation, and tumor mass). These results suggest that aspirin's chemopreventive effects may be due, in part, to the drug blocking the proneoplastic action of platelets, and the potential use of Aspirin-PC/PL2200 as an effective and safer chemopreventive agent for colorectal cancer and possibly other cancers. Cancer Prev Res; 10(2); 142-52. ©2016 AACR.


Assuntos
Aspirina/farmacologia , Neoplasias do Colo/patologia , Ciclo-Oxigenase 1/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/enzimologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C
3.
Mol Cancer Ther ; 15(12): 2894-2904, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27638860

RESUMO

To determine the efficacy of a novel and safer (for gastrointestinal tract) aspirin (aspirin-PC) in preclinical models of ovarian cancer, in vitro dose-response studies were performed to compare the growth-inhibitory effect of aspirin-PC versus aspirin on three human (A2780, SKOV3ip1, and HeyA8) and a mouse (ID8) ovarian cancer cell line over an 8-day culture period. In the in vivo studies, the aspirin test drugs were studied alone and in the presence of a VEGF-A inhibitor (bevacizumab or B20), due to an emerging role for platelets in tumor growth following antiangiogenic therapy, and we examined their underlying mechanisms. Aspirin-PC was more potent (vs. aspirin) in blocking the growth of both human and mouse ovarian cancer cells in monolayer culture. Using in vivo model systems of ovarian cancer, we found that aspirin-PC significantly reduced ovarian cancer growth by 50% to 90% (depending on the ovarian cell line). The efficacy was further enhanced in combination with Bevacizumab or B20. The growth-inhibitory effect on ovarian tumor mass and number of tumor nodules was evident, but less pronounced for aspirin and the VEGF inhibitors alone. There was no detectable gastrointestinal toxicity. Both aspirin and aspirin-PC also inhibited cell proliferation, angiogenesis, and increased apoptosis of ovarian cancer cells. In conclusion, PC-associated aspirin markedly inhibits the growth of ovarian cancer cells, which exceeds that of the parent drug, in both cell culture and in mouse model systems. We also found that both aspirin-PC and aspirin have robust antineoplastic action in the presence of VEGF-blocking drugs. Mol Cancer Ther; 15(12); 2894-904. ©2016 AACR.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Aspirina/farmacologia , Neovascularização Patológica , Neoplasias Ovarianas/patologia , Fosfatidilcolinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Tromboxanos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Physiol Rep ; 4(6)2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27033447

RESUMO

NSAIDuse is limited due to the drugs' toxicity to the gastrointestinal mucosa, an action incompletely understood. Lower gut injury induced byNSAIDs is dependent on bile secretion and is reported to increase the growth of a number of bacterial species, including an enterococcal species,Enterococcus faecalis This study examined the relationships between indomethacin (INDO)-induced intestinal injury/bleeding, small bowel overgrowth (SBO) and dissemination of enterococci, and the contribution of bile secretion to these pathological responses. Rats received either a sham operation (SO) or bile duct ligation (BDL) prior to administration of two daily subcutaneous doses of saline orINDO, and 24 h later, biopsies of ileum and liver were collected for plating on selective bacterial media. Fecal hemoglobin (Hb) and blood hematocrit (Hct) were measured to assess intestinal bleeding. Of the four treatment groups, onlySO/INDOrats experienced a significant 10- to 30-fold increase in fecal Hb and reduction in Hct, indicating thatBDLattenuatedINDO-induced intestinal injury/bleeding. Ileal enterococcal colony-forming units were significantly increased (500- to 1000-fold) inSO/INDOrats. Of all groups, only theSO/INDOrats demonstrated gut injury, and this was associated with enterococcal overgrowth of the gut and dissemination to the liver. We also demonstrated thatINDO-induced intestinal injury andE. faecalisovergrowth was independent of the route of administration of the drug, as similar findings were observed in rats orally dosed with theNSAID Bile secretion plays an important role inINDO-induced gut injury and appears to support enterococcal overgrowth of the intestine.NSAID-induced enterococcalSBOmay be involved either as a compensatory response to gut injury or with the pathogenic process itself and the subsequent development of sepsis.


Assuntos
Anti-Inflamatórios não Esteroides , Ductos Biliares/metabolismo , Bile/metabolismo , Enterococcus faecalis/crescimento & desenvolvimento , Hemorragia Gastrointestinal/microbiologia , Doenças do Íleo/microbiologia , Íleo/microbiologia , Indometacina , Animais , Translocação Bacteriana , Ductos Biliares/cirurgia , Modelos Animais de Doenças , Enterococcus faecalis/metabolismo , Fezes/química , Hemorragia Gastrointestinal/induzido quimicamente , Hemorragia Gastrointestinal/metabolismo , Hemorragia Gastrointestinal/patologia , Hemoglobinas/metabolismo , Doenças do Íleo/induzido quimicamente , Doenças do Íleo/metabolismo , Doenças do Íleo/patologia , Íleo/metabolismo , Íleo/patologia , Ligadura , Fígado/microbiologia , Masculino , Ratos Sprague-Dawley
5.
Am J Physiol Gastrointest Liver Physiol ; 308(9): G785-93, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25721304

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) are widely used to treat a number of conditions, and proton pump inhibitors (PPIs) are often used to prevent NSAID-induced gastric mucosal damage; however, the effects of NSAIDs on intestinal motility are poorly understood. The purpose of the present study is to determine the effects of a prototypical NSAID, indomethacin, either alone or in conjunction with the PPI omeprazole, on intestinal motility. Rats were randomly divided into four groups treated with vehicle, omeprazole, indomethacin, or a combination of indomethacin and omeprazole. Intestinal motility and transit were measured along with inflammatory mediators in the intestinal smooth muscle, markers of mucosal damage, and bacterial counts in the intestinal wall. Indomethacin, but not omeprazole, caused mucosal injury indicated by lower gut bleeding; however, both omeprazole and indomethacin suppressed contractile activity and frequency in the distal part of the small intestine. Cotreatment with omeprazole did not reduce indomethacin-induced intestinal bleeding. Furthermore, although indomethacin caused increased inflammation as indicated by increased edema development and inflammatory mediators, cotreatment with omeprazole did not reduce inflammation in the intestinal smooth muscle or prevent the increased bacterial count in the intestinal wall induced by indomethacin. We conclude that both NSAID and PPI treatment suppressed contractile activity in the distal regions of the small intestine. The suppression of intestinal contractility was associated with increased inflammation in both cases; however, indomethacin and omeprazole appear to affect intestinal motility by different mechanisms.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Motilidade Gastrointestinal/efeitos dos fármacos , Íleo/efeitos dos fármacos , Indometacina/toxicidade , Jejuno/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Omeprazol/toxicidade , Inibidores da Bomba de Prótons/toxicidade , Animais , Biomarcadores/metabolismo , Enterite/induzido quimicamente , Enterite/metabolismo , Enterite/fisiopatologia , Hemorragia Gastrointestinal/induzido quimicamente , Hemorragia Gastrointestinal/patologia , Hemorragia Gastrointestinal/fisiopatologia , Íleo/metabolismo , Íleo/microbiologia , Íleo/patologia , Íleo/fisiopatologia , Mediadores da Inflamação/metabolismo , Jejuno/metabolismo , Jejuno/microbiologia , Jejuno/patologia , Jejuno/fisiopatologia , Masculino , Músculo Liso/metabolismo , Músculo Liso/fisiopatologia , Ratos Sprague-Dawley
6.
Am J Physiol Gastrointest Liver Physiol ; 308(3): G217-22, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25477376

RESUMO

Indomethacin is a powerful analgesic nonsteroidal anti-inflammatory drug (NSAID), but is limited in use by its primary side effect to cause gastrointestinal bleeding and serious injury. One factor important for exacerbating NSAID injury is the presence of bile acids, which may interact with indomethacin to form toxic mixed micelles in the gut. The development of a safer gastrointestinal formulation of indomethacin that is chemically complexed with phosphatidylcholine (PC-indomethacin) may offer an improved therapeutic agent, particularly in the presence of bile acid, but its potential protective mechanism is incompletely understood. Intestinal epithelial cells (IEC-6) were tested for injury with indomethacin (alone and plus various bile acids) compared with PC-indomethacin (alone and plus bile acids). To explore a role for bile acid uptake into cells as a requirement for NSAID injury, studies were performed using Madin-Darby canine kidney cells transfected with the apical sodium-dependent bile acid transporter (ASBT). Indomethacin, but not PC-indomethacin, was directly and dose-dependently injurious to IEC-6 cells. Similarly, the combination of any bile acid plus indomethacin, but not PC-indomethacin, induced cell injury. The expression of ASBT had a modest effect on the acute cytotoxicity of indomethacin in the presence of some conjugated bile acids. Complexing PC with indomethacin protected against the acute intestinal epithelial injury caused by indomethacin regardless of the presence of bile acids. The presence of luminal bile acid, but not its carrier-mediated uptake into the enterocyte, is required for acute indomethacin-induced cell injury. It is likely that initial cell damage induced by indomethacin occurs at or near the cell membrane, an effect exacerbated by bile acids and attenuated by PC.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Ácidos e Sais Biliares/toxicidade , Hemorragia Gastrointestinal/tratamento farmacológico , Indometacina/toxicidade , Fosfatidilcolinas/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Hemorragia Gastrointestinal/induzido quimicamente , Camundongos , Transportadores de Ânions Orgânicos Dependentes de Sódio/efeitos dos fármacos , Simportadores/efeitos dos fármacos
7.
J Biol Chem ; 288(50): 35660-70, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24165125

RESUMO

Eukaryotic cell membranes are organized into functional lipid and protein domains, the most widely studied being membrane rafts. Although rafts have been associated with numerous plasma membrane functions, the mechanisms by which these domains themselves are regulated remain undefined. Bile acids (BAs), whose primary function is the solubilization of dietary lipids for digestion and absorption, can affect cells by interacting directly with membranes. To investigate whether these interactions affected domain organization in biological membranes, we assayed the effects of BAs on biomimetic synthetic liposomes, isolated plasma membranes, and live cells. At cytotoxic concentrations, BAs dissolved synthetic and cell-derived membranes and disrupted live cell plasma membranes, implicating plasma membrane damage as the mechanism for BA cellular toxicity. At subtoxic concentrations, BAs dramatically stabilized domain separation in Giant Plasma Membrane Vesicles without affecting protein partitioning between coexisting domains. Domain stabilization was the result of BA binding to and disordering the nonraft domain, thus promoting separation by enhancing domain immiscibility. Consistent with the physical changes observed in synthetic and isolated biological membranes, BAs reorganized intact cell membranes, as evaluated by the spatial distribution of membrane-anchored Ras isoforms. Nanoclustering of K-Ras, related to nonraft membrane domains, was enhanced in intact plasma membranes, whereas the organization of H-Ras was unaffected. BA-induced changes in Ras lateral segregation potentiated EGF-induced signaling through MAPK, confirming the ability of BAs to influence cell signal transduction by altering the physical properties of the plasma membrane. These observations suggest general, membrane-mediated mechanisms by which biological amphiphiles can produce their cellular effects.


Assuntos
Ácido Desoxicólico/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Lipossomos/química , Lipossomos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Micelas , Nanoestruturas/química , Ratos , Solubilidade , Proteínas ras/química , Proteínas ras/metabolismo
8.
Gut Liver ; 7(1): 7-15, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23423874

RESUMO

The mucosa of the gastrointestinal (GI) tract exhibits hydrophobic, nonwettable properties that protect the underlying epithelium from gastric acid and other luminal toxins. These biophysical characteristics appear to be attributable to the presence of an extracellular lining of surfactant-like phospholipids on the luminal aspects of the mucus gel layer. Phosphatidylcholine (PC) represents the most abundant and surface-active form of gastric phospholipids. PC protected experimental rats from a number of ulcerogenic agents and/or conditions including nonsteroidal anti-inflammatory drugs (NSAIDs), which are chemically associated with PC. Moreover, preassociating a number of the NSAIDs with exogenous PC prevented a decrease in the hydrophobic characteristics of the mucus gel layer and protected rats against the injurious GI side effects of NSAIDs while enhancing and/or maintaining their therapeutic activity. Bile plays an important role in the ability of NSAIDs to induce small intestinal injury. NSAIDs are rapidly absorbed from the GI tract and, in many cases, undergo enterohepatic circulation. Thus, NSAIDs with extensive enterohepatic cycling are more toxic to the GI tract and are capable of attenuating the surface hydrophobic properties of the mucosa of the lower GI tract. Biliary PC plays an essential role in the detoxification of bile salt micelles. NSAIDs that are secreted into the bile injure the intestinal mucosa via their ability to chemically associate with PC, which forms toxic mixed micelles and limits the concentration of biliary PC available to interact with and detoxify bile salts. We have worked to develop a family of PC-associated NSAIDs that appear to have improved GI safety profiles with equivalent or better therapeutic efficacy in both rodent model systems and pilot clinical trials.

9.
J Surg Res ; 180(1): 140-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23158407

RESUMO

BACKGROUND: The gastric mucosa is protected in part by a hydrophobic layer of phosphatidylcholine (PC) that overlies the mucus gel on the stomach. Endotoxin treatment (i.e., lipopolysaccharide [LPS]) results in an apparent disruption of this layer, as evidenced by a reduction in surface hydrophobicity and an increase in transmural permeability. The current studies compared PC and lyso-PC levels in mucus and gastric mucosa before and after LPS treatment, and examined potential mechanisms for surface phospholipid changes. METHODS: Rats were administered LPS (5 mg//kg, intraperitoneally) and samples were collected after 5 h for analysis of PC and its primary degradant, lyso-PC, in the loosely and firmly adherent mucus layers and the mucosa. The dependence of LPS-induced effects on gastric alkalinization, PC synthetic activity, and intestinal reflux material was assessed. RESULTS: The gastric contents after LPS, which also contained duodenal reflux material, had greatly increased amounts of PC and lyso-PC. The firmly adherent mucus layer was unchanged. The gastric mucosa after LPS revealed significant reductions of PC levels and no change in lyso-PC content. These phospholipid changes were not caused by alkalinization of the stomach or altered PC synthesis. Prevention of duodenogastric reflux by pylorus ligation blocked the LPS-induced increase in luminal lyso-PC and the reduction in mucosal PC. CONCLUSIONS: LPS appears to induce a release of PC from gastric mucosa into the lumen, along with degradation of PC to lyso-PC, without an effect on PC synthesis. Component(s) of intestinal reflux material appear to be required for these effects. The lowered PC levels in gastric mucosa after LPS may contribute to reduced barrier properties of this tissue.


Assuntos
Mucosa Gástrica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Fosfatidilcolinas/metabolismo , Animais , Mucosa Gástrica/metabolismo , Ligadura , Ratos , Ratos Sprague-Dawley
10.
Biochim Biophys Acta ; 1818(12): 3040-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22885171

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are frequently used to treat chronic pain and inflammation. However, prolonged use of NSAIDs has been known to result in Gastrointestinal (GI) ulceration/bleeding, with a bile-mediated mechanism underlying their toxicity to the lower gut. Bile acids (BAs) and phosphatidylcholines (PCs), the major components of bile, form mixed micelles to reduce the membrane disruptive actions of monomeric BAs and simple BA micelles. NSAIDs are suspected to alter the BA/PC balance in the bile, but the molecular interactions of NSAID-BA or NSAID-BA-PC remain undetermined. In this work, we used a series of all-atom molecular dynamics simulations of cholic acid (CA), ibuprofen (IBU) and dodecylphosphocholine (DPC) mixtures to study the spontaneous aggregation of CA and IBU as well as their adsorption on a DPC micelle. We found that the size of CA-IBU mixed micelles varies with their molar ratio in a non-linear manner, and that micelles of different sizes adopt similar shapes but differ in composition and internal interactions. These observations are supported by NMR chemical shift changes, NMR ROESY crosspeaks between IBU and CA, and dynamic light scattering experiments. Smaller CA-IBU aggregates were formed in the presence of a DPC micelle due to the segregation of CA and IBU away from each other by the DPC micelle. While the larger CA-IBU aggregates arising from higher IBU concentrations might be responsible for NSAID-induced intestinal toxicity, the absence of larger CA-IBU aggregates in the presence of DPC micelles may explain the observed attenuation of NSAID toxicity by PCs.


Assuntos
Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/toxicidade , Ácido Cólico/química , Ibuprofeno/química , Micelas , Fosforilcolina/análogos & derivados , Anti-Inflamatórios não Esteroides/farmacologia , Ácidos e Sais Biliares/química , Ibuprofeno/toxicidade , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Fosforilcolina/química
11.
Digestion ; 86(2): 171-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22907267

RESUMO

BACKGROUND/AIMS: Proton pump inhibitors (PPIs) are widely used to prevent nonsteroidal anti-inflammatory drug (NSAID)-induced peptic ulcers. NSAIDs produce small intestinal injury and some PPIs have been reported to protect against NSAID-induced small bowel injury in rats. The aim of this study was to compare PPIs, revaprazan, and phosphatidylcholine-associated indomethacin (Indo-PC) for protection against indomethacin (Indo)-induced small bowel injury. METHODS: Rat intestinal epithelial cells (IEC-6) were pretreated with omeprazole, lansoprazole, or revaprazan prior to exposure to Indo or Indo-PC. Cell viability was assessed by methyl thiazolyl tetrazolium assay. Omeprazole, lansoprazole, or revaprazan was administered orally to rats prior to the vehicle or Indo. Indo-PC was administered alone. After 24 h, small intestinal erosions were counted; intestinal bleeding was assessed as the hemoglobin concentration of small intestinal fluid. RESULTS: Omeprazole, lansoprazole, and revaprazan did not protect against Indo-induced IEC-6 cell injury. Indo-PC was less damaging in vitro than Indo alone. In vivo, neither omeprazole nor lansoprazole protected against Indo-induced small bowel injury; however, revaprazan pretreatment and Indo-PC resulted in significantly fewer erosions (>50% reduction) or bleeding (>80% reduction). CONCLUSION: PPIs showed no small bowel protective effect in vitro or in vivo. Revaprazan showed a small bowel protective effect in vivo, whereas Indo-PC was protective both in vitro and in vivo.


Assuntos
2-Piridinilmetilsulfinilbenzimidazóis/farmacologia , Anti-Inflamatórios não Esteroides/efeitos adversos , Hemorragia Gastrointestinal , Indometacina/efeitos adversos , Intestino Delgado/efeitos dos fármacos , Omeprazol/farmacologia , Fosfatidilcolinas/farmacologia , Inibidores da Bomba de Prótons/farmacologia , Pirimidinonas/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Hemorragia Gastrointestinal/induzido quimicamente , Hemorragia Gastrointestinal/prevenção & controle , Mucosa Intestinal/efeitos dos fármacos , Lansoprazol , Masculino , Ratos , Ratos Sprague-Dawley
12.
Biochim Biophys Acta ; 1821(7): 994-1002, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22521764

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) are one of the most widely consumed pharmaceuticals, yet both the mechanisms involved in their therapeutic actions and side-effects, notably gastrointestinal (GI) ulceration/bleeding, have not been clearly defined. In this study, we have used a number of biochemical, structural, computational and biological systems including; Fourier Transform InfraRed (FTIR). Nuclear Magnetic Resonance (NMR) and Surface Plasmon Resonance (SPR) spectroscopy, and cell culture using a specific fluorescent membrane probe, to demonstrate that NSAIDs have a strong affinity to form ionic and hydrophobic associations with zwitterionic phospholipids, and specifically phosphatidylcholine (PC), that are reversible and non-covalent in nature. We propose that the pH-dependent partition of these potent anti-inflammatory drugs into the phospholipid bilayer, and possibly extracellular mono/multilayers present on the luminal interface of the mucus gel layer, may result in profound changes in the hydrophobicity, fluidity, permeability, biomechanical properties and stability of these membranes and barriers. These changes may not only provide an explanation of how NSAIDs induce surface injury to the GI mucosa as a component in the pathogenic mechanism leading to peptic ulceration and bleeding, but potentially an explanation for a number of (COX-independent) biological actions of this family of pharmaceuticals. This insight also has proven useful in the design and development of a novel class of PC-associated NSAIDs that have reduced GI toxicity while maintaining their essential therapeutic efficacy to inhibit pain and inflammation.


Assuntos
Anti-Inflamatórios não Esteroides/química , Aspirina/química , Mucosa Gástrica/efeitos dos fármacos , Ibuprofeno/química , Naproxeno/química , Fosfatidilcolinas/química , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Corantes Fluorescentes , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ibuprofeno/farmacologia , Bicamadas Lipídicas/química , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Naproxeno/farmacologia , Compostos de Piridínio , Espectroscopia de Infravermelho com Transformada de Fourier , Ressonância de Plasmônio de Superfície
13.
Antimicrob Agents Chemother ; 54(9): 3618-24, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20547807

RESUMO

Diarrhea is one of the most common infirmities affecting international travelers, occurring in 20 to 50% of persons from industrialized countries visiting developing regions. Enterotoxigenic Escherichia coli (ETEC) is the most common causative agent and is isolated from approximately half of the cases of traveler's diarrhea. Rifaximin, a largely water-insoluble, nonabsorbable (<0.4%) antibiotic that inhibits bacterial RNA synthesis, is approved for use for the treatment of traveler's diarrhea caused by diarrheagenic E. coli. However, the drug has minimal effect on the bacterial flora or the infecting E. coli strain in the aqueous environment of the colon. The purpose of the present study was to evaluate the antimicrobial effect and bioavailability of rifaximin in aqueous solution in the presence and absence of physiologic concentrations of bile acids. The methods used included growth measurement of ETEC (strain H10407), rifaximin solubility measurements, total bacterial protein determination, and assessment of the functional activity of rifaximin by monitoring inhibition of bacterial beta-galactosidase expression. Solubility studies showed rifaximin to be 70- to 120-fold more soluble in bile acids (approximately 30% in 4 mM bile acids) than in aqueous solution. Addition of both purified bile acids and human bile to rifaximin at subinhibitory and inhibitory concentrations significantly improved the drug's anti-ETEC effect by 71% and 73%, respectively, after 4 h. This observation was confirmed by showing a decrease in the overall amount of total bacterial protein expressed during incubation of rifaximin plus bile acids. Rifaximin-treated samples containing bile acids inhibited the expression of ETEC beta-galactosidase at a higher magnitude than samples that did not contain bile acids. The study provides data showing that bile acids solubilize rifaximin on a dose-response basis, increasing the drug's bioavailability and antimicrobial effect. These observations suggest that rifaximin may be more effective in the treatment of infections in the small intestine, due to the higher concentration of bile in this region of the gastrointestinal tract than in the colon. The water insolubility of rifaximin is the likely explanation for the drug's minimal effects on colonic flora and fecal pathogens, despite in vitro susceptibility.


Assuntos
Antibacterianos/farmacologia , Ácidos e Sais Biliares/química , Rifamicinas/farmacologia , Antibacterianos/química , Escherichia coli Enterotoxigênica/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Rifamicinas/química , Rifaximina , Solubilidade
14.
Am J Physiol Gastrointest Liver Physiol ; 298(5): G722-31, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20203063

RESUMO

The injurious effect of nonsteroidal anti-inflammatory drugs (NSAIDs) in the small intestine was not appreciated until the widespread use of capsule endoscopy. Animal studies found that NSAID-induced small intestinal injury depends on the ability of these drugs to be secreted into the bile. Because the individual toxicity of amphiphilic bile acids and NSAIDs directly correlates with their interactions with phospholipid membranes, we propose that the presence of both NSAIDs and bile acids alters their individual physicochemical properties and enhances the disruptive effect on cell membranes and overall cytotoxicity. We utilized in vitro gastric AGS and intestinal IEC-6 cells and found that combinations of bile acid, deoxycholic acid (DC), taurodeoxycholic acid, glycodeoxycholic acid, and the NSAID indomethacin (Indo) significantly increased cell plasma membrane permeability and became more cytotoxic than these agents alone. We confirmed this finding by measuring liposome permeability and intramembrane packing in synthetic model membranes exposed to DC, Indo, or combinations of both agents. By measuring physicochemical parameters, such as fluorescence resonance energy transfer and membrane surface charge, we found that Indo associated with phosphatidylcholine and promoted the molecular aggregation of DC and potential formation of larger and isolated bile acid complexes within either biomembranes or bile acid-lipid mixed micelles, which leads to membrane disruption. In this study, we demonstrated increased cytotoxicity of combinations of bile acid and NSAID and provided a molecular mechanism for the observed toxicity. This mechanism potentially contributes to the NSAID-induced injury in the small bowel.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Ácidos e Sais Biliares/metabolismo , Indometacina/efeitos adversos , Fosfolipídeos/metabolismo , Animais , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Ácido Desoxicólico/farmacologia , Interações Medicamentosas , Transferência Ressonante de Energia de Fluorescência , Ácido Glicodesoxicólico/farmacologia , Humanos , Intestino Delgado/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Lipossomos/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Permeabilidade , Fosfolipídeos/farmacologia , Ratos , Ácido Taurodesoxicólico/farmacologia
15.
Shock ; 33(6): 634-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19940811

RESUMO

Endotoxemia from sepsis can injure the gastrointestinal tract through mechanisms that have not been fully elucidated. We have shown that LPS induces an increase in gastric permeability in parallel with the luminal appearance of secretory phospholipase A2 (sPLA2) and its product, lysophosphatidylcholine (lyso-PC). We proposed that sPLA2 acted on the gastric hydrophobic barrier, composed primarily of phosphatidylcholine (PC), to degrade it and produce lyso-PC, an agent that is damaging to the mucosa. In the present study, we have tested whether lyso-PC and/or sPLA2 have direct damaging effects on the hydrophobic barriers of synthetic and mucosal surfaces. Rats were administered LPS (5 mg/kg, i.p.), and gastric contents were collected 5 h later for analysis of sPLA2 and lyso-PC content. Using these measured concentrations, direct effects of sPLA2 and lyso-PC were determined on (a) surface hydrophobicity as detected with an artificial PC surface and with intact gastric mucosa (contact angle analysis) and (b) cell membrane disruption of gastric epithelial cells (AGS). Both lyso-PC and sPLA2 increased significantly in the collected gastric juice of LPS-treated rats. Using similar concentrations to the levels in gastric juice, the contact angle of PC-coated slides declined after incubation with either pancreatic sPLA2 or lyso-PC. Similarly, gastric contact angles seen in control rats were significantly decreased in sPLA2 and lyso-PC-treated rats. In addition, we observed dose-dependent injurious effects of both lyso-PC and sPLA2 in gastric AGS cells. An LPS-induced increase in sPLA2 activity in the gastric lumen and its product, lyso-PC, are capable of directly disrupting the gastric hydrophobic layer and may contribute to gastric barrier disruption and subsequent inflammation.


Assuntos
Mucosa Gástrica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Lisofosfatidilcolinas/farmacologia , Fosfolipases A2 Secretórias/metabolismo , Animais , Venenos de Abelha/farmacologia , Endotoxemia/metabolismo , Células Epiteliais/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lipopolissacarídeos/toxicidade , Masculino , Ratos , Ratos Sprague-Dawley , Venenos de Serpentes/farmacologia , Células Tumorais Cultivadas
16.
Shock ; 30(6): 729-33, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18496240

RESUMO

The hydrophobic surface layer of the gastrointestinal (GI) tract, which has been attributed to the presence of phosphatidylcholine (PC) in the mucus gel, protects the mucosa of the GI tract and is disrupted by parenteral LPS treatment. We investigated the potential for repletion of this layer as a means to prevent LPS-induced GI injury. Rats were treated orally with PC 1 h before LPS (i.p.). Gastric and ileal tissues were assessed for changes in permeability 5 h later, and gastric fluid was analyzed for signs of GI-related LPS effects (bile acid reflux, increased volume, and pH) and gastric injury (bleeding). Serum TNF-alpha was assessed as a measure of a non-GI, LPS response. Radiolabeled PC was tracked through the GI tract to verify the extent of luminal exposure during the time of the study. Pretreatment with oral PC significantly blocked permeability increases in gastric and ileal tissue due to LPS. A portion of orally administered PC gained access to the entire GI tract in 1 h. Exogenous PC did not prevent the increase in serum TNF-alpha or gastric fluid volume or pH induced by LPS, nor did it prevent the duodenogastric reflux of bile acid. There was a tendency for PC to reduce gastric bleeding after LPS. Orally administered PC seems to act directly on the mucosa to prevent GI permeability disturbances due to LPS. Under the conditions studied, oral PC does not block systemic effects of LPS. However, enteral formulations containing PC may be useful adjuncts in the prevention of GI injury from endotoxemia.


Assuntos
Mucosa Gástrica/imunologia , Inflamação/prevenção & controle , Mucosa Intestinal/imunologia , Lipopolissacarídeos/farmacologia , Fosfatidilcolinas/administração & dosagem , Fosfatidilcolinas/farmacologia , Administração Oral , Animais , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Concentração de Íons de Hidrogênio , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa
17.
J Trauma ; 64(3): 681-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18332808

RESUMO

BACKGROUND: Gastric function in trauma patients is poorly understood. In animals, shock causes gastric luminal alkalinization and bile reflux. In trauma patients, studies of stress gastritis prophylaxis demonstrated with continuous gastric pH monitoring that the stomach became alkaline even without antisecretory therapy. Therefore, we hypothesized that trauma patients have an alkaline gastric environment that may be because of bile reflux. METHODS: A prospective observational study at an urban Level I trauma center was performed. All major torso trauma patients (severe head injuries excluded) who met the criteria for standardized shock resuscitation were eligible for inclusion. A 12.5 Fr silastic pH probe (Sandhill Scientific) was placed in the stomach and the gastric pH continuously monitored for 7 days. Patients received no stress gastritis prophylaxis. Gastric samples were obtained each day and assayed for total bile acids and pH. RESULTS: Twelve patients were entered into the study. Mean age was 31 years +/- 4 years, 67% men, 75% blunt mechanism of injury, and mean Injury Severity Score 28 +/- 3. Three patients (25%) developed multiple organ failure and four acquired ventilator-associated pneumonia. During the first day of continuous pH monitoring, 9 of 12 patients had a gastric pH >4 for the majority of the day with 7 patients having essentially no acid production. During subsequent days, gastric pH began to drop and by the 4th day the majority of each day was spent at a pH <4. Additionally, gastric pH of patients with ventilator-associated pneumonia or multiple organ failure tended to be more alkaline. Bile acid was present in the gastric fluid of all patients in varying amounts. However, there was no significant correlation between gastric pH and bile acid concentration. CONCLUSIONS: Traumatic injury causes gastric luminal alkalinization that may be related, only in part, to bile acid reflux. Other alkalinizing factors remain to be elucidated.


Assuntos
Ácido Gástrico/metabolismo , Ressuscitação/métodos , Traumatismos Torácicos/complicações , Equilíbrio Ácido-Base , Adulto , Idoso , Análise de Variância , Feminino , Determinação da Acidez Gástrica , Refluxo Gastroesofágico/etiologia , Humanos , Concentração de Íons de Hidrogênio , Escala de Gravidade do Ferimento , Análise dos Mínimos Quadrados , Masculino , Pessoa de Meia-Idade , Monitorização Fisiológica , Estudos Prospectivos , Sistema de Registros , Fatores de Risco
18.
Shock ; 30(2): 206-11, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18180698

RESUMO

A hydrophobic layer of phosphatidylcholine (PC) overlies and protects the surface of the gastrointestinal (GI) tract, contributing to barrier integrity. During critical illness such as sepsis, gut barrier integrity is compromised, which could be related to degradation of PC. The purpose of this study was to investigate a role for luminal (secretory) phospholipase A2 (sPLA(2)) in LPS-induced GI injury. Rats were treated with LPS (5 mg/kg) or saline for 0.5, 1, 3, and 5 h. The gastric and ileal luminal contents were collected for determination of sPLA(2) activity, and the luminal lipids were analyzed using thin layer chromatography for lyso-PC content. The GI permeability was assessed in vivo with fluorescein-isothiocyanate dextran 4000 and rats were tested with or without a specific sPLA(2) inhibitor. LPS induced significant increases in sPLA(2) activity and lyso-PC content in the gastric and ileal lumens at 5 h. In addition, LPS treated rats showed a significant increase in GI permeability to fluorescein-isothiocyanate dextran in both the stomach and ileum at 5 h, which was prevented by pretreatment with the sPLA(2) inhibitor. In response to LPS, sPLA(2) activity increases in the GI tract lumen where it may degrade the extracellular protective phospholipid layer and membranes, producing injurious lyso-PC and increased GI permeability. Pretreatment with an orally active sPLA(2) inhibitor blocks the LPS-induced increase in GI permeability, and may suggest a new approach to fortify the GI mucosal barrier and prevent complications from endotoxin in trauma and other patients.


Assuntos
Endotoxemia/enzimologia , Endotoxemia/fisiopatologia , Mucosa Gástrica/enzimologia , Mucosa Gástrica/fisiopatologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/fisiopatologia , Lipopolissacarídeos/toxicidade , Fosfolipases A2 Secretórias/fisiologia , Animais , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Mucosa Gástrica/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Masculino , Permeabilidade , Fosfatidilcolinas/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia
19.
J Trauma ; 64(1): 105-10, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18188106

RESUMO

BACKGROUND: The pathogenesis of multiple organ failure (MOF) in trauma patients may involve the gastrointestinal tract, but its exact origins remain elusive. In a prospective study, the gastric fluid of major torso trauma patients was examined for evidence of duodenogastric reflux and potential gastric injury, and was compared with patient outcomes regarding MOF. METHODS: Patient samples were collected daily for 4 days by nasogastric tube and analyzed for pH, hemoglobin, and bile acid. Blood was collected for analysis of C-reactive protein (CRP). Outcomes were recorded for the presence or absence of MOF. RESULTS: The results showed that most patients exhibited alkaline gastric contents (pH >/=4.9) and elevated levels of hemoglobin immediately after the trauma. Although non-MOF patients demonstrated a decline of both mean gastric pH and bleeding by day 4, MOF patients maintained significant elevations in pH during this time period. Mean total bile acid levels were increased in all patients, signifying the presence of duodenogastric reflux. However, there were no clear differences in mean bile acid concentrations between MOF and non-MOF patients over time, although MOF patients tended to exhibit higher levels. All patients showed a progressive rise in serum CRP during the first 24 hours after trauma, which was maintained for 4 days. The initial rise in serum CRP in MOF patients was delayed compared with that in non-MOF patients. CONCLUSIONS: We conclude that duodenogastric reflux occurs in trauma patients in the first few days after trauma and may contribute to elevated gastric pH and bleeding. Further study is needed to verify whether monitoring the gastric juice of trauma patients during the first several days of hospitalization, for alkaline pH and excessive blood in the gastric lumen, could lead to better assessments of patient status.


Assuntos
Refluxo Duodenogástrico/etiologia , Hemorragia Gastrointestinal/etiologia , Insuficiência de Múltiplos Órgãos/etiologia , Ferimentos e Lesões/complicações , Adulto , Refluxo Duodenogástrico/diagnóstico , Feminino , Determinação da Acidez Gástrica , Suco Gástrico/fisiologia , Hemorragia Gastrointestinal/diagnóstico , Humanos , Concentração de Íons de Hidrogênio , Masculino , Observação , Estudos Prospectivos , Choque Hemorrágico/complicações , Ferimentos e Lesões/fisiopatologia
20.
J Gastroenterol Hepatol ; 23(8 Pt 2): e384-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18086111

RESUMO

BACKGROUND AND AIMS: A mechanism for protection of gastrointestinal (GI) and hepatic cells from damaging detergent actions of bile acids appears to involve the bile component, phosphatidylcholine (PC). Non-steroidal anti-inflammatory drugs (NSAIDs) induce intestinal injury in direct proportion to their ability to be excreted into bile, and are known to chemically associate with PC. We investigated the role of bile acids and PC in the mechanism of indomethacin-induced epithelial injury. METHODS: Rats were injected orally or intravenously with radiolabeled indomethacin and their bile was collected over time for determination of NSAID secretion. Bile from rats treated with or without indomethacin was used in studies of red blood cell (RBC) hemolysis as a measure of membrane cytotoxicity. The bile salt, sodium deoxycholate (SDC), and indomethacin were tested alone and in combination with PC on RBC and on hepatic HepG2 cells. RESULTS: Intravenously or orally given indomethacin was quantitatively excreted (approximately 50%) into bile over a 2-h study period. Bile from a rat treated with indomethacin or bile with exogenous indomethacin was cytotoxic to RBC, and the injury was prevented by the addition of PC. Hepatocytes exposed to SDC showed injury that could be dose-dependently prevented by PC, and reversed by indomethacin. CONCLUSIONS: Biliary PC plays an important physiological role in protecting GI and hepatic epithelia from the cytotoxic actions of bile salts. The ability of NSAIDs excreted into the bile to associate with mixed bile salt micelles and reduce the protective action of the PC may be a critical component in the drugs' pathogenic mechanism.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Sistema Biliar/metabolismo , Doença Hepática Induzida por Substâncias e Drogas , Gastroenteropatias/induzido quimicamente , Indometacina/farmacocinética , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Indometacina/efeitos adversos , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...