Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cells ; 10(6)2021 05 21.
Article in English | MEDLINE | ID: mdl-34064187

ABSTRACT

Once known exclusively for their role in nutrients absorption, bile acids have emerged as signaling molecules, generated from cholesterol breakdown, acting on several immune cells by activating a variety of receptors including the G protein-coupled bile acid receptor 1 (GPABR1 or TGR5), the Farnesoid-X-receptor (FXR) and, as recently discovered, the retinoid-related orphan receptors (ROR)γt. GPBAR1, FXR, and RORγt are highly expressed in cells of the innate and adaptive immune system (i.e., dendritic cells (DCs), macrophages, innate lymphoid 3 cells (ILC3s), and T helper 17 (Th17) lymphocytes) and plays an important role in regulating intestinal and liver immunity, highlighting a role for various bile acid species in regulating immune responses to intestinal microbial antigens. While primary bile acids are generated from the cholesterol breakdown secondary bile acids, the GPBAR1 ligands, and oxo-bile acids derivatives, the RORγt ligands, are generated by the intestinal microbiota, highlighting the potential of these bile acids in mediating the chemical communication between the intestinal microbiota and the host. Changes in intestinal microbiota, dysbiosis, alter the composition of the bile acid pool, promoting the activation of the immune system and development of chronic inflammation. In this review, we focus on the molecular mechanisms by which an altered bile acid signaling promotes intestinal inflammation.


Subject(s)
Bile Acids and Salts , Gastrointestinal Microbiome/immunology , Immunity, Innate , Inflammatory Bowel Diseases , Animals , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Receptors, Cytoplasmic and Nuclear/immunology , Receptors, G-Protein-Coupled/immunology
2.
Dig Dis Sci ; 66(3): 674-693, 2021 03.
Article in English | MEDLINE | ID: mdl-33289902

ABSTRACT

Bile acids are a group of chemically different steroids generated at the host/microbial interface. Indeed, while primary bile acids are the end-product of cholesterol breakdown in the host liver, secondary bile acids are the products of microbial metabolism. Primary and secondary bile acids along with their oxo derivatives have been identified as signaling molecules acting on a family of cell membrane and nuclear receptors collectively known as "bile acid-activated receptors." Members of this group of receptors are highly expressed throughout the gastrointestinal tract and mediate the bilateral communications of the intestinal microbiota with the host immune system. The expression and function of bile acid-activated receptors FXR, GPBAR1, PXR, VDR, and RORγt are highly dependent on the structure of the intestinal microbiota and negatively regulated by intestinal inflammation. Studies from gene ablated mice have demonstrated that FXR and GPBAR1 are essential to maintain a tolerogenic phenotype in the intestine, and their ablation promotes the polarization of intestinal T cells and macrophages toward a pro-inflammatory phenotype. RORγt inhibition by oxo-bile acids is essential to constrain Th17 polarization of intestinal lymphocytes. Gene-wide association studies and functional characterizations suggest a potential role for impaired bile acid signaling in development inflammatory bowel diseases (IBD). In this review, we will focus on how bile acids and their receptors mediate communications of intestinal microbiota with the intestinal immune system, describing dynamic changes of bile acid metabolism in IBD and the potential therapeutic application of targeting bile acid signaling in these disorders.


Subject(s)
Bile Acids and Salts/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Signal Transduction/immunology , Bile Acids and Salts/immunology , Gastrointestinal Microbiome/immunology , Humans , Immune System Phenomena/physiology , Intestinal Mucosa/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
Sci Rep ; 6: 30802, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27492684

ABSTRACT

Targeted disruption of leukocyte trafficking to the gut represents a promising approach for the treatment of inflammatory bowel diseases (IBDs). CCR5, the shared receptor for MIP1α and ß and RANTES, is expressed by multiple leukocytes. Here, we aimed to determine the role of CCR5 in mediating leukocyte trafficking in models of colitis, and evaluate the therapeutic potential of maraviroc, an orally active CCR5 antagonist used in the treatment of CCR5-tropic HIV. Acute and chronic colitis were induced by administration of DSS or TNBS to wild-type and CCR5(-/-) mice or adoptive transfer of splenic naïve CD4(+) T-cells from wild type or CCR5(-/-) mice into RAG-1(-/-). CCR5 gene ablation reduced the mucosal recruitment and activation of CCR5-bearing CD4(+) and CD11b(+) leukocytes, resulting in profound attenuation of signs and symptoms of inflammation in the TNBS and transfer models of colitis. In the DSS/TNBS colitis and in the transfer model, maraviroc attenuated development of intestinal inflammation by selectively reducing the recruitment of CCR5 bearing leukocytes. In summary, CCR5 regulates recruitment of blood leukocytes into the colon indicating that targeting CCR5 may offer therapeutic options in IBDs.


Subject(s)
Chemokine CCL5/metabolism , Colitis/pathology , Inflammation/prevention & control , Intestinal Mucosa/metabolism , Animals , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chemokines/antagonists & inhibitors , Colitis/chemically induced , Colon/physiology , Cyclohexanes/pharmacology , Cyclohexanes/therapeutic use , Dextran Sulfate/toxicity , Disease Models, Animal , Homeodomain Proteins/genetics , Inflammation/immunology , Inflammation/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Maraviroc , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/cytology , Th17 Cells/immunology , Triazoles/pharmacology , Triazoles/therapeutic use , Trinitrobenzenesulfonic Acid/toxicity , Up-Regulation/drug effects
4.
J Gastroenterol ; 50(7): 707-19, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25708288

ABSTRACT

Bile acids are the end product of cholesterol metabolism. Synthesized in the liver, primary bile acids are secreted by hepatocytes and are transformed by intestinal microbiota into secondary bile acids. In addition to their role in cholesterol and lipid absorption, bile acids act as signaling molecules activating a family of nuclear and G-protein-coupled receptors collectively known as bile acid activated receptors (BARs). These receptors are expressed at high density in enterohepatic tissues, but their expression occurs throughout the body and their activation mediates regulatory functions of bile acids on lipids and glucose metabolism and immunity. In the gastrointestinal tract, BARs maintain intestinal integrity, and their deletion makes the intestine more susceptible to the damage caused by acetylsalicylic acid and nonsteroidal anti-inflammatory drugs (NSAIDs). Deficiency in farnesoid X receptor and G-protein-coupled bile acid receptor 1 genes alters the expression/activity of cystathione γ-lyase and endothelial nitric oxide synthase, two genes involved in the synthesis of hydrogen sulfide and nitric oxide, i.e., two gaseous mediators that have been shown to be essential in maintaining the intestinal homeostasis. In addition, farnesoid X receptor regulates the expression of transporters required for secretion of phospholipid by hepatocytes. Because phospholids attenuate intestinal injury caused by acetylsalicylic acid and NSAIDs, BAR agonism could be exploited to protect the intestinal mucosa against injury caused by anti-inflammatory medications. This approach might be useful in the prevention of so-called NSAID enteropathy, a common clinical condition occurring in long-term users of NSAIDs, which is not effectively prevented either by cotreatment with proton pump inhibitors or by the use of coxibs.


Subject(s)
Bile Acids and Salts/metabolism , Intestinal Mucosa/metabolism , Lipid Metabolism , Homeostasis , Humans , Signal Transduction
5.
Eur J Pharmacol ; 723: 194-201, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24315932

ABSTRACT

The protease inhibitor ritonavir is part of the highly active anti-retroviral therapy (HAART) successfully used in the treatment of human immunodeficiency virus (HIV)-1 infection. There is evidence that ritonavir alters intestinal permeability and induces damage to the small intestine. Because HIV infected patients taking HAART are at high risk for developing cardiovascular complications, there might be a need for the use of low dose of aspirin (ASA) to prevent ischemic events. Similarly, long term survival exposes HIV infected persons to detrimental interactions of ritonavir with non-steroidal anti-inflammatory drugs (NSAIDs). In the present work we tested whether ritonavir worsens intestinal injury caused by NSAIDs and ASA. C57BL6 mice were treated for 25 days with ritonavir and for a further 5 days with the combination of ritonavir plus ASA or ritonavir plus naproxen. In a second set of experiments C57BL6 mice were cotreated with ritonavir plus misoprostol, a PGE1 analog. We found that ritonavir administration caused intestinal damage and its co-administration with naproxen or ASA exacerbated the severity of injury and intestinal inflammation, as assessed by measuring haematocrit, MPO, mucosal levels of PGE2 and mRNA levels of iNOS, MCP-1 and VLA-1. Co-administration of misoprostol protected against intestinal damage induced by naproxen and ritonavir. In conclusion we demonstrated that ritonavir causes intestinal damage and that its association with NSAIDs or ASA worsens the damage caused by COX-inhibitors. Misoprostol rescues from intestinal damage caused by ritonavir. Further studies are need to clarify whether this observation has a clinical readout.


Subject(s)
Cyclooxygenase Inhibitors/adverse effects , HIV Protease Inhibitors/adverse effects , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Ritonavir/adverse effects , Alprostadil/pharmacology , Animals , Apoptosis/drug effects , Aspirin/adverse effects , Chemokine CCL2/genetics , Dinoprostone/metabolism , Dinoprostone/pharmacology , Drug Synergism , HT29 Cells , Humans , Integrin alpha1beta1/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Ketoprofen/adverse effects , Mice , Mice, Inbred C57BL , Misoprostol/pharmacology , Naproxen/adverse effects , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/adverse effects
6.
Br J Pharmacol ; 168(1): 225-37, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22881598

ABSTRACT

BACKGROUND AND PURPOSE: Low doses of aspirin (acetylsalicylic acid; ASA) and non-steroidal anti-inflammatory drugs (NSAIDs) increase the risk of gastrointestinal bleeding. GPBAR1 is a bile acid receptor expressed in the gastrointestinal tract. Here, we have investigated whether GPBAR1 was required for mucosal protection in models of gastrointestinal injury caused by ASA and NSAIDs. EXPERIMENTAL APPROCH: GPBAR1(+/+) and GPBAR1(-/-) mice were given ASA (10-50 mg.kg(-1)) or naproxen. Gastric and intestinal mucosal damage was assessed by measuring lesion scores. KEY RESULTS: Expression of GPBAR1, mRNA and protein, was detected in mouse stomach. Mice lacking GPBAR1 were more sensitive to gastric and intestinal injury caused by ASA and NSAIDs and exhibited a markedly reduced expression of cystathionine-γ-liase (CSE), cystathionine-ß-synthase (CBS) and endothelial NOS enzymes required for generation of H(2)S and NO, in the stomach. Treating GPBAR1(+/+) mice with two GPBAR1 agonists, ciprofloxacin and betulinic acid, rescued mice from gastric injury caused by ASA and NSAIDs. The protective effect of these agents was lost in GPBAR1(-/-) mice. Inhibition of CSE by DL-propargylglycine completely reversed protection afforded by ciprofloxacin in wild type mice, whereas treating mice with an H(2)S donor restored the protective effects of ciprofloxacin in GPBAR1(-/-) mice. Deletion of GPBAR1 altered the morphology of the small intestine and increased sensitivity to injury caused by naproxen. CONCLUSION AND IMPLICATIONS: GPBAR1 is essential to maintain gastric and intestinal mucosal integrity. GPBAR1 agonists protect against gastrointestinal injury caused by ASA and NSAIDs by a COX-independent mechanism.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Aspirin/adverse effects , Gastric Mucosa/metabolism , Gastrointestinal Hemorrhage/chemically induced , Gastrointestinal Hemorrhage/prevention & control , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Bile/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastrointestinal Hemorrhage/pathology , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/pathology , Male , Mice , Naproxen/adverse effects
7.
Br J Pharmacol ; 164(8): 1929-38, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21564085

ABSTRACT

BACKGROUND AND PURPOSE: Low doses of acetyl salicylic acid (ASA) and non-steroidal anti-inflammatory drugs (NSAIDs) cause gastrointestinal damage. The farnesoid X receptor (FXR) is a bile acid sensor essential for maintenance of intestinal homeostasis. Here, we have investigated whether FXR is required for mucosal protection in models of gastrointestinal injury caused by ASA and NSAIDs and if FXR activation has potential in the treatment or prevention of gastrointestinal injury caused by these agents. EXPERIMENTAL APPROACH: FXR(+/+) and FXR(-/-) mice were given ASA (10 to 100 mg·kg(-1) ) or NSAIDs. Gastric and intestinal mucosal damage assessed by measuring lesion scores. FXR were activated by giving mice natural (chenodeoxycholic acid; CDCA) or synthetic (GW4064) FXR agonists. KEY RESULTS: FXR, mRNA and protein, was detected in human and mouse stomach. FXR(-/-) mice were more prone to develop severe gastric and intestinal injury in response to ASA and NSAIDs and showed a severe reduction in the gastrointestinal expression of cystathionine-γ-lyase (CSE), an enzyme required for generation of hydrogen sulphide. CSE expression was reduced by ≈50% in wild-type mice challenged with ASA. Treating wild-type mice but not FXR(-/-) mice with CDCA or GW4064 protected against gastric injury caused by ASA and NSAIDs, by a CSE-dependent and cycloxygenase- and NO-independent, mechanism. FXR activation by GW4064 rescued mice from intestinal injury caused by naproxen. CONCLUSIONS AND IMPLICATIONS: FXR was essential to maintain gastric and intestinal mucosal barriers. FXR agonists protected against gastric injury caused by ASA and NSAIDs by a CSE-mediated mechanism.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Gastrointestinal Tract/injuries , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/metabolism , Homeostasis , Humans , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Inflamm Allergy Drug Targets ; 10(2): 133-40, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21275896

ABSTRACT

Nonsteroidal anti-inflammatory pain medications, commonly referred to as NSAIDs, are effective treatment for pain, fever and inflammation. However their use associates with a 4-6 fold increase in the risk of gastrointestinal bleeding. The basic mode of action of NSAIDs lies in the inhibition of cyclooxygenases (COXs), a family of enzymes involved in the generation of prostaglandins (PGs). The COX exists at least in two isoforms, COX-1 and COX-2, with PGs mediating inflammation at site of injury generated by the COX-2, while COX-1 produces PGs that are essential in maintaining integrity in the gastrointestinal tract. Selective inhibitors of COX-2, the coxibs, spare the gastrointestinal tract while exerting anti-inflammatory and analgesic effects. However, their use has been linked to an increased risk of thrombo-embolic events. Nitric oxide (NO) and hydrogen sulfide (H(2)S), are potent vasodilatory agents that maintain mucosal integrity in the gastrointestinal tract. In the last decade hybrid molecules that release NO or H(2)S have been coupled with non-selective NSAIDs to generate new classes of anti-inflammatory and analgesic agents with the potential to spare the gastrointestinal and cardiovascular system. These agents, the NO-releasing NSAIDs, or CINOD, and the H(2)S-releasing NSAIDs are currently investigated as a potential alternative to NSAIDs and coxibs. Naproxcinod has been the first, and so far the only, CINOD extensively investigated in clinical trials. Despite its promising profile, the approval of this drug was recently rejected by the Food and Drug Administration because the lack of long-term controlled studies. NSAIDs that release H(2)S as a mechanism to support an enhanced gastrointestinal and cardiovascular safety are being investigated in preclinical studies. Either naproxen or diclofenac coupled to an H(2)S releasing moiety has been reported to cause less gastrointestinal and cardiovascular injury than parent NSAIDs in preclinical models.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Hydrogen Sulfide/metabolism , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cyclooxygenase Inhibitors/adverse effects , Drug Design , Gastrointestinal Hemorrhage/chemically induced , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Nitric Oxide/metabolism , Pain/drug therapy , Pain/physiopathology
9.
J Immunol ; 183(10): 6657-66, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19880446

ABSTRACT

Immune-mediated liver diseases including autoimmune and viral hepatitis are a major health problem worldwide. In this study, we report that activation of the farnesoid X receptor (FXR), a member of the ligand-activated nuclear receptor superfamily and bile sensor highly expressed in the liver, attenuates liver injury in a model of autoimmune hepatitis induced by Con A. We found that FXR gene ablation results in a time-dependent increase of liver expression (up to 20-fold in a 9-mo-old mouse) of osteopontin, a NKT cell-derived extracellular matrix protein and immunoregulatory cytokine. In comparison to wild-type, FXR(-/-) mice are more susceptible to Con A-induced hepatitis and react to Con A administration by an unregulated production of osteopontin. Administering wild-type mice with a synthetic FXR agonist attenuated Con A-induced liver damage and liver expression of the osteopontin gene. By in vitro studies, we found that FXR is expressed by primarily isolated NKT cells and its ablation favors ostepontin production in response to Con A. Chromatin immunoprecipitation assay and coimmunoprecipitation experiments demonstrate that the short heterodimer partner (SHP), a nuclear receptor and FXR target, was expressed by NKT cell hybridomas and increased in response to FXR activation. FXR activates SHP that interacts with and inhibits c-Jun binding to the osteopontin promoter. These data indicate that in NKT cells, FXR activation causes a SHP-mediated inhibition of osteopontin production. These data support the notion that the bile acid sensor FXR regulates the activation of liver NKT cells.


Subject(s)
Hepatitis/immunology , Natural Killer T-Cells/immunology , Osteopontin/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Acute Disease , Animals , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Cell Line , Concanavalin A/pharmacology , Hepatitis/genetics , Hepatitis/metabolism , Interferon-gamma/agonists , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/agonists , Interleukin-4/immunology , Interleukin-4/metabolism , Liver/immunology , Liver/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogens/pharmacology , Natural Killer T-Cells/metabolism , Osteopontin/agonists , Osteopontin/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/immunology , RNA, Messenger/drug effects , RNA, Messenger/immunology , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
10.
Gastroenterology ; 136(2): 530-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18996377

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel diseases are relatively common diseases of the gastrointestinal tract. The relative therapeutic efficacy of glucocorticoids used in inflammatory bowel diseases resides in part in their capability to inhibit activity of nuclear factor kappaB (NF-kappaB), a transcription factor central to the inflammatory process, and the consequent production of T-helper 1 (Th1)-type cytokines. Previous studies indicate that increased expression in transgenic mice of glucocorticoid-induced leucine zipper (GILZ), a gene rapidly induced by glucocorticoids, inhibits NF-kappaB and Th1 activity. METHODS: We performed experiments with the aim to test the susceptibility of GILZ transgenic (GILZ-TG) mice to dinitrobenzene sulfonic acid-induced colitis. RESULTS: Consistent with a decreased Th1 response, GILZ-TG mice were less susceptible to colitis induction as compared with wild-type littermates, while they were more susceptible to Th2-mediated colitis. The inhibition was comparable to that obtained with dexamethasone treatment. Moreover, diminished intestinal tissue damage, associated with inhibition of NF-kappaB nuclear translocation, interferon-gamma, tumor necrosis factor-alpha, and interleukin-1 production in CD4+ T lymphocytes of the lamina propria, was evident in GILZ-TG as compared with wild-type mice. In addition, inhibition of colitis development was also evident when GILZ fusion protein was delivered in vivo in dinitrobenzene sulfonic acid-treated WT animals as well as in interleukin-10 knockout mice. CONCLUSIONS: Together these results demonstrate that GILZ mimics the effects of glucocorticoids, suggesting a contribution of this protein to the anti-inflammatory activity of glucocorticoids in Th1-induced colitis.


Subject(s)
Colitis/metabolism , Colitis/pathology , Th1 Cells/metabolism , Th1 Cells/pathology , Transcription Factors/metabolism , Animals , Colitis/chemically induced , Colon/metabolism , Colon/pathology , Dinitrofluorobenzene/analogs & derivatives , Disease Models, Animal , Genetic Predisposition to Disease , Glucocorticoids/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-1/metabolism , Leucine Zippers/drug effects , Mice , Mice, Knockout , Mice, Transgenic , NF-kappa B/metabolism , Oxazolone , Th2 Cells/metabolism , Th2 Cells/pathology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Hepatology ; 47(2): 659-67, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18098324

ABSTRACT

UNLABELLED: Increased intrahepatic resistance in cirrhotic livers is caused by endothelial dysfunction and impaired formation of two gaseous vasodilators, nitric oxide (NO) and hydrogen sulfide (H(2)S). Homocysteine, a sulfur-containing amino acid and H(2)S precursor, is formed from hepatic methionine metabolism. In the systemic circulation, hyperhomocystenemia impairs vasodilation and NO production from endothelial cells. Increased blood levels of homocysteine are common in patients with liver cirrhosis. In this study, we demonstrate that acute liver perfusion with homocysteine impairs NO formation and intrahepatic vascular relaxation induced by acetylcholine in methoxamine-precontracted normal livers (7.3% +/- 3.0% versus 26% +/- 2.7%; P < 0.0001). In rats with mild, diet-induced hyperhomocystenemia, the vasodilating activity of acetylcholine was markedly attenuated, and incremental increases in flow induced a greater percentage of increases in perfusion pressure than in control livers. Compared with normal rats, animals rendered cirrhotic by 12 weeks' administration of carbon tetrachloride exhibited a greater percentage of increments in perfusion pressure in response to shear stress (P < 0.05), and intrahepatic resistance to incremental increases in flow was further enhanced by homocysteine (P < 0.05). In normal hyperhomocysteinemic and cirrhotic rat livers, endothelial dysfunction caused by homocysteine was reversed by perfusion of the livers with sodium sulfide. Homocysteine reduced NO release from sinusoidal endothelial cells and also caused hepatic stellate cell contraction; this suggests a dual mechanism of action, with the latter effect being counteracted by H(2)S. CONCLUSION: Impaired vasodilation and hepatic stellate cell contraction caused by homocysteine contribute to the dynamic component of portal hypertension.


Subject(s)
Homocysteine/pharmacology , Hydrogen Sulfide/pharmacology , Liver Circulation/physiology , Liver/physiology , Methionine/physiology , Microcirculation/physiology , Acetylcholine/pharmacology , Animals , Disease Models, Animal , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/physiopathology , Liver/drug effects , Liver Circulation/drug effects , Liver Cirrhosis/blood , Liver Cirrhosis/physiopathology , Male , Microcirculation/drug effects , Rats , Rats, Wistar
12.
Anticancer Drugs ; 18(9): 1081-91, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17704659

ABSTRACT

The use of epirubicin is limited by the risk of a dilatory congestive heart failure that develops as a consequence of induction of a mitochondrial-dependent cardiomyocyte apoptosis. In a previous in-vitro study, we have provided evidence that a new formulation of pegylated epirubicin- bearing moieties that release nitric oxide, named BP-747, exerted a potent antitumoral activity against a colon cancer cell line, which was completely devoid of cytotoxic activity against cardiomyocytes. The aim of this study was to investigate the antitumoral and cardiotoxic profile of BP-747 in Caco-2 and SKOV-2 tumor-bearing mice. Epirubicin-induced cardiomyopathy was detected by clinical (survival, weight loss), anatomical (heart weight loss) and biochemical evaluations (measurement of serum troponin and creatine phosphokinase levels). The antitumoral activity was investigated by the measurement of tumor diameters and weight. In comparison with free epirubicin and pegylated epirubicin, BP-747 showed more potent antineoplastic effects, as demonstrated by the 95% reduction of tumor volume. Moreover, while administration of epirubicin and pegylated epirubicin resulted in the development of a severe anthracycline cardiomyopathy, BP-747-treated mice were virtually devoid of clinical and biochemical signs of cardiotoxicity. The present data provide evidence that addition of a nitric oxide-releasing moiety to pegylated epirubicin confers a new and unique cytotoxic profile to the drug.


Subject(s)
Antibiotics, Antineoplastic , Cardiomyopathies/chemically induced , Epirubicin/analogs & derivatives , Heart/drug effects , Neoplasms/drug therapy , Polyethylene Glycols , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Cardiomyopathies/enzymology , Cardiomyopathies/pathology , Caspases/metabolism , Cell Line, Tumor , Creatine Kinase/blood , Epirubicin/adverse effects , Epirubicin/chemistry , Epirubicin/therapeutic use , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Neoplasm Transplantation , Polyethylene Glycols/adverse effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Troponin/blood , Xenograft Model Antitumor Assays
13.
Trends Mol Med ; 13(7): 298-309, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17588816

ABSTRACT

The farnesoid X receptor (FXR) is a metabolic nuclear receptor expressed in the liver, intestine, kidney and adipose tissue. By regulating the expression and function of genes involved in bile acid (BA) synthesis, uptake and excretion, FXR has emerged as a key gene involved in the maintenance of cholesterol and BA homeostasis. FXR ligands are currently under clinical investigation for the treatment of cholestasis, dyslipidemic disorders and conditions of insulin resistance in type 2 diabetes and non-alcoholic steatohepatitis (NASH). Because activation of FXR impacts a considerable number of genes, development of FXR modulators that selectively regulate specific pathways will limit potentially undesirable side effects. Interaction of FXR with other BAs and xenobiotics sensors such as the constitutive androstane receptor and the pregnane X receptor might allow the development of combination therapies for liver and metabolic disorders.


Subject(s)
DNA-Binding Proteins/agonists , Liver Diseases/drug therapy , Metabolic Diseases/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Atherosclerosis/metabolism , Bile Acids and Salts/metabolism , Cholestasis/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Ligands , Liver/metabolism , Liver Diseases/metabolism , Metabolic Diseases/metabolism , Models, Biological , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
14.
FASEB J ; 20(6): 765-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16455756

ABSTRACT

The use of the anthracycline epirubicin (EPI) is limited by the risk of a dilatory congestive heart failure that develops as a consequence of induction of a mitochondrial-dependent cardiomyocyte and endothelial cell apoptosis. Nitric oxide (NO) increases the antitumoral activity of several chemotherapics, while it provides protection against apoptosis induced by oxidative stress both in endothelial cells and cardiomyocytes. The aim of the present study was to investigate whether the addition of an NO-releasing moiety to a pegylated derivative of EPI (p-EPI-NO) confers to the drug a different cytotoxic profile against tumoral and normal cells. The cytotoxic profile of the drugs was investigated in Caco-2 cell line, in embryonic rat heart-derived myoblasts (H9c2), in adult cardiomyocytes, and in endothelial cells (HUVEC). p-EPI-NO was more efficient than EPI in inducing Caco-2 cell apoptosis, while it spared HUVEC, H9c2 cells and adult cardiomyocytes from EPI-induced toxicity. Exposure of cells to p-EPI-NO resulted in a NO-mediated inhibition of cellular respiration followed by mitochondrial membrane depolarization and cell death in Caco-2 cells but not in HUVEC and H9c2 cells in which mitochondrial membrane polarization was maintained at the expense of glycolytically generated ATP. These findings indicate that addition of an NO-releasing moiety to p-EPI increases the anti-neoplastic activity of the drug, while it reduces its cytotoxicity against nonneoplastic cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Epirubicin/analogs & derivatives , Nitric Oxide/metabolism , Polyethylene Glycols/pharmacology , Adenosine Triphosphate/metabolism , Animals , Caco-2 Cells , Cells, Cultured , Electron Transport Complex IV/metabolism , Epirubicin/pharmacology , Humans , Mice , Myocytes, Cardiac/metabolism , Oxygen Consumption , Rats
15.
Gastroenterology ; 128(5): 1243-57, 2005 May.
Article in English | MEDLINE | ID: mdl-15887108

ABSTRACT

BACKGROUND & AIMS: Uncontrolled T-cell activation plays a critical role in the pathogenesis of inflammatory bowel diseases. Therefore, pharmacological strategies directed toward restoring the normal responsiveness of the immune system could be effective in the treatment of these pathologic conditions. The addition of a nitric oxide-releasing moiety to conventional drugs, such as aspirin and other anti-inflammatory analgesic drugs, results in new chemical entities with potent immunomodulatory activities. The aim of this study was to investigate the immunomodulatory activity of a nitric oxide-releasing derivative of mesalamine (NCX-456), as compared with standard mesalamine, in 2,4,6-trinitrobenzene sulfonic acid-induced colitis in mice. METHODS: Cells and tissues from mice with 2,4,6-trinitrobenzene sulfonic acid-induced colitis and from interleukin 10-deficient mice with spontaneous chronic colitis receiving treatment with several doses of NCX-456 or mesalamine were analyzed for morphology, cytokine production, and apoptosis. RESULTS: NCX-456, but not mesalamine, administration resulted in a marked reduction in clinical, histological, and immunologic signs of colitis in both models. NCX-456 inhibited the release of T-helper type 1-derived cytokines and increased the release of the regulatory T cell-derived cytokines interleukin 10 and transforming growth factor beta. In vitro analyses showed that NCX-456 inhibited proliferation and caused selective apoptosis of the subset of activated lamina propria T-helper type 1 cells, whereas it was ineffective for regulatory T-cell function and survival. CONCLUSIONS: Collectively, these data show that NCX-456 inhibits lamina propria T-helper type 1 function and stimulates the activity of interleukin 10- and transforming growth factor beta-secreting cells, thus restoring mucosal immune homeostasis and suppressing intestinal inflammation.


Subject(s)
Aminosalicylic Acids/pharmacology , Apoptosis/drug effects , Colitis/drug therapy , Colitis/immunology , Nitric Oxide/metabolism , T-Lymphocyte Subsets/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/immunology , Cells, Cultured , Colitis/chemically induced , Colon/cytology , Colon/immunology , Immunologic Factors/pharmacology , In Vitro Techniques , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism
16.
Proc Natl Acad Sci U S A ; 100(19): 10937-41, 2003 Sep 16.
Article in English | MEDLINE | ID: mdl-12960371

ABSTRACT

In addition to inhibiting cyclooxygenase (COX)-1-derived prostanoid biosynthesis, aspirin acetylates COX-2, enabling the conversion of arachidonic acid to 15(R)-epi lipoxin A4, or aspirin-triggered lipoxin (ATL). Selective COX-2 inhibitors block ATL formation and exacerbate mucosal injury in rats treated with aspirin. In the present study, we have examined whether inhibition of COX-2 activity in healthy volunteers taking aspirin exacerbates gastric mucosal injury and if such an effect would be prevented by NCX-4016, a NO-releasing derivative of aspirin. Thirty-two volunteers were randomized to receive 2 wk of treatment with NCX-4016 (800 mg twice a day) or aspirin (100 mg once a day) alone or in combination with 200 mg of celecoxib twice a day. Mucosal damage was assessed by endoscopy. The mean mucosal injury score was 5.8 +/- 1.8 in subjects treated with aspirin and 2.4 +/- 0.7 (P < 0.01 vs. aspirin) in subjects treated with NCX-4016. Administration of celecoxib increased the injury score in volunteers treated with aspirin (9.9 +/- 1.9) but not in subjects taking NCX-4016 (1.5 +/- 0.8). Aspirin and NCX-4016 caused a comparable suppression of serum thromboxane B2 levels and increased urinary excretion of ATL. Celecoxib inhibited endotoxin-induced prostaglandin E2 generation in whole blood by approximately 80% and abolished ATL formation. These findings suggests that (i) aspirin and NCX-4016 trigger ATL formation in humans, (ii) celecoxib inhibits ATL formation and exacerbates the mucosal injury caused by low doses of aspirin, and (iii) the NO-donating moiety of NCX-4016 protects the gastric mucosa even in the presence of suppression of COX-1 and COX-2.


Subject(s)
Aspirin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Gastric Mucosa/drug effects , Isoenzymes/drug effects , Prostaglandin-Endoperoxide Synthases/drug effects , Sulfonamides/pharmacology , Adult , Aspirin/analogs & derivatives , Celecoxib , Female , Gastric Mucosa/enzymology , Humans , Lipoxins/metabolism , Male , Pyrazoles
17.
Gastroenterology ; 124(5): 1381-94, 2003 May.
Article in English | MEDLINE | ID: mdl-12730878

ABSTRACT

BACKGROUND & AIMS: Uncontrolled T-cell activation plays a critical role in the pathogenesis of inflammatory bowel diseases. Therefore, pharmacologic strategies directed to restore the normal responsiveness of the immune system by deleting inappropriately activated T cells could be efficacious in the treatment of these pathologic conditions. Galectin-1 is an endogenous lectin expressed in lymphoid organs that plays a role in the maintenance of central and peripheral tolerance. The aim of the present study was to evaluate the therapeutic effects of galectin-1 on T-helper cell type 1-mediated experimental colitis induced by intrarectal administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) in mice. METHODS: Cells and tissues from mice with TNBS colitis receiving treatment with several doses of human recombinant galectin-1 (hrGAL-1) were analyzed for morphology, cytokine production, and apoptosis. RESULTS: Prophylactic and therapeutic administration of rhGAL-1 resulted in a striking improvement in the clinical and histopathologic aspects of the disease. hrGAL-1 reduced the number of hapten-activated spleen T cells, decreased inflammatory cytokine production, and profoundly reduced the ability of lamina propria T cells to produce IFN gamma in vitro. Moreover, hrGAL-1 led to the appearance of apoptotic mononuclear cells in colon tissue when administered in vivo and induced selective apoptosis of TNBS-activated lamina propria T cells in vitro. CONCLUSION: Collectively, these data show that hrGAL-1 exerts protective and immunomodulatory activity in TNBS-induced colitis and it might be effective in the treatment of inflammatory bowel diseases.


Subject(s)
Colitis/drug therapy , Galectin 1/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Colitis/immunology , Colitis/pathology , Disease Models, Animal , Galectin 1/genetics , Gene Expression/physiology , Interferon-gamma/metabolism , Interleukin-1/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Spleen/cytology , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism
18.
Gastroenterology ; 124(3): 600-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612897

ABSTRACT

BACKGROUND AND AIMS: NCX-4016 is a nitric oxide-releasing derivative of aspirin with antiplatelet activity. The aim of this study was to investigate the effect of NCX-4016 on gastrointestinal mucosa and platelet functions in healthy human volunteers. METHODS: This was a parallel-group, double-blind, placebo-controlled study. Forty healthy subjects were randomly allocated to receive 7 days of treatment with NCX-4016 (400 and 800 mg twice daily), equimolar doses of aspirin (200 and 420 mg twice daily), or placebo. Upper endoscopies were performed before and at the end of the treatment period, and gastroduodenal lesions were graded using a predefined scoring system. Basal and posttreatment platelet aggregation in response to arachidonic acid (AA) and serum thromboxane (TX) B(2) and AA-stimulated platelet TXB(2) production were investigated. RESULTS: Mucosal endoscopic injury score on day 7 was 0.63 +/- 0.16 in the placebo group and 11.0 +/- 3.0 and 16.1 +/- 1.6 in healthy volunteers treated with 200 and 420 mg aspirin twice daily (P < 0.0001 vs. placebo). NCX-4016 was virtually devoid of gastric and duodenal toxicity, resulting in a total gastric and duodenal endoscopic score of 1.38 +/- 0.3 and 1.25 +/- 0.5 (P < 0.0001 vs. aspirin, not significant vs. placebo). NCX-4016 inhibited AA-induced platelet aggregation as well as serum TXB(2) and platelet TXB(2) generation induced by AA to the same extent as aspirin (not significant vs. aspirin). CONCLUSIONS: In this study, we have proven the concept that addition of an NO-donating moiety to aspirin results in a new chemical entity that maintains cyclooxygenase-1 and platelet inhibitory activity while nearly avoiding gastrointestinal damage.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/analogs & derivatives , Aspirin/pharmacology , Cardiotonic Agents/pharmacology , Digestive System/drug effects , Adult , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Aspirin/adverse effects , Cardiotonic Agents/adverse effects , Digestive System/pathology , Double-Blind Method , Endoscopy, Gastrointestinal , Female , Gastric Mucosa/drug effects , Humans , Intestinal Mucosa/drug effects , Male , Pilot Projects , Placebos/adverse effects , Placebos/pharmacology , Platelet Aggregation/drug effects , Thromboxane B2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...