Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Biochem Pharmacol ; 218: 115900, 2023 12.
Article in English | MEDLINE | ID: mdl-37926268

ABSTRACT

While patients with nonalcoholic fatty liver disease (NAFLD) are at increased risk to develop clinically meaningful cardiovascular diseases (CVD), there are no approved drug designed to target the liver and CVD component of NAFLD. GPBAR1, also known as TGR5, is a G protein coupled receptor for secondary bile acids. In this study we have investigated the effect of GPBAR1 activation by BAR501, a selective GPBAR1 agonist, in Apolipoprotein E deficient (ApoE-/-) mice fed a high fat diet and fructose (Western diet), a validated model of NAFLD-associated atherosclerosis. Using aortic samples from patients who underwent surgery for abdominal aneurism, and ex vivo experiments with endothelial cells and human macrophages, we were able to co-localize the expression of GPBAR1 in CD14+ and PECAM1+ cells. Similar findings were observed in the aortic plaques from ApoE-/- mice. Treating ApoE-/- mice with BAR501, 30 mg/kg for 14 weeks, attenuated the body weight gain while ameliorated the insulin sensitivity by increasing the plasma concentrations of GLP-1 and FGF15. Activation of GPBAR1 reduced the aorta thickness and severity of atherosclerotic lesions and decreased the amount of plaques macrophages. Treating ApoE-/- mice reshaped the aortic transcriptome promoting the expression of anti-inflammatory genes, including IL-10, as also confirmed by tSNE analysis of spleen-derived macrophages. Feeding ApoE-/- mice with BAR501 redirected the bile acid synthesis and the composition of the intestinal microbiota. In conclusion, GPBAR1 agonism attenuates systemic inflammation and improve metabolic profile in a genetic/dietetic model of atherosclerosis. BAR501 might be of utility in the treatment for NAFLD-related CVD.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Apolipoproteins E , Atherosclerosis/drug therapy , Cardiovascular Diseases/complications , Disease Models, Animal , Endothelial Cells , Inflammation/drug therapy , Inflammation/complications , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Receptors, G-Protein-Coupled/genetics
2.
J Am Heart Assoc ; 12(23): e031241, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37996988

ABSTRACT

BACKGROUND: Patients with nonalcoholic fatty liver disease are at increased risk to develop atherosclerotic cardiovascular diseases. FXR and GPBAR1 are 2 bile acid-activated receptors exploited in the treatment of nonalcoholic fatty liver disease: whether dual GPBAR1/FXR agonists synergize with statins in the treatment of the liver and cardiovascular components of nonalcoholic fatty liver disease is unknown. METHODS AND RESULTS: Investigations of human aortic samples obtained from patients who underwent surgery for aortic aneurysms and Gpbar1-/-, Fxr-/-, and dual Gpbar1-/-Fxr-/- mice demonstrated that GPBAR1 and FXR are expressed in the aortic wall and regulate endothelial cell/macrophage interactions. The expression of GPBAR1 in the human endothelium correlated with the expression of inflammatory biomarkers. Mice lacking Fxr and Gpbar1-/-/Fxr-/- display hypotension and aortic inflammation, along with altered intestinal permeability that deteriorates with age, and severe dysbiosis, along with dysregulated bile acid synthesis. Vasomotor activities of aortic rings were altered by Gpbar1 and Fxr gene ablation. In apolipoprotein E-/- and wild-type mice, BAR502, a dual GPBAR1/FXR agonist, alone or in combination with atorvastatin, reduced cholesterol and low-density lipoprotein plasma levels, mitigated the development of liver steatosis and aortic plaque formation, and shifted the polarization of circulating leukocytes toward an anti-inflammatory phenotype. BAR502/atorvastatin reversed intestinal dysbiosis and dysregulated bile acid synthesis, promoting a shift of bile acid pool composition toward FXR antagonists and GPBAR1 agonists. CONCLUSIONS: FXR and GPBAR1 maintain intestinal, liver, and cardiovascular homeostasis, and their therapeutic targeting with a dual GPBAR1/FXR ligand and atorvastatin holds potential in the treatment of liver and cardiovascular components of nonalcoholic fatty liver disease.


Subject(s)
Bile Acids and Salts , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Bile Acids and Salts/metabolism , Dysbiosis/complications , Dysbiosis/metabolism , GTP-Binding Proteins/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Receptors, G-Protein-Coupled/metabolism
3.
Sci Total Environ ; 884: 163846, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37137373

ABSTRACT

Integrated Multitrophic Aquaculture (IMTA) seems to be one of the best solutions for sustainable aquaculture. Within the Remedia LIFE Project, an experimental IMTA plant was put in place in the Mar Grande of Taranto (Mediterranean Sea, Southern Italy). The polyculture of several bioremediating organisms, such as mussels, tubeworms, sponges, and seaweeds, was combined with a coastal cage fish farm, in order to remove organic and inorganic wastes coming from the fish's metabolism. To verify the effectiveness of the system, the ex ante measurement of chemical-physical variables, trophic status, microbial contamination, and zoobenthos community health was compared with the results of the same measurement performed one year and two years after the implementation of the experimental IMTA plant. The results were encouraging, since a reduction in total nitrogen concentration in the seawater (from 43.4 ± 8.9 to 5.6 ± 3.7 µM/l), a reduction in microbial pollution indicators in the seawater (total coliforms: from 280 ± 18 MPN/100 mL to 0; E. coli: from 33 ± 1.3 MPN/100 mL to 0) and in the sediments (total coliforms: from 230 ± 6.2 MPN/100 g to 170 ± 9; E. coli: from 40 ± 9.4 MPN/100 g to 0), an enhancement of the trophic status (TRIX: from 4.45 ± 1.29 to 3.84 ± 0.18), and an increase in the zoobenthic quality indices and biodiversity were recorded (AMBI: from 4.8 to 2.4; M-AMBI: from 0.14 to 0.7). These results prove that the Remedia LIFE project's purpose was achieved. The selected bioremediators worked synergistically, improving water and sediments quality within the fish farm area. Moreover, bioremediating organisms increased their weight as a result of wastes uptake, producing, as co-products, large amounts of additional biomass. This could be commercially exploited, thus being an added value of the IMTA plant. Based on our findings, the promotion of eco-friendly practices to ameliorate ecosystem health should be encouraged.


Subject(s)
Bivalvia , Ecosystem , Animals , Escherichia coli , Quality Improvement , Seawater , Aquaculture , Environmental Monitoring
4.
Front Oncol ; 13: 1140730, 2023.
Article in English | MEDLINE | ID: mdl-36998446

ABSTRACT

Introduction: The leukemia inhibitory factor (LIF), is a cytokine belonging to IL-6 family, whose overexpression correlate with poor prognosis in cancer patients, including pancreatic ductal adenocarcinoma (PDAC). LIF signaling is mediate by its binding to the heterodimeric LIF receptor (LIFR) complex formed by the LIFR receptor and Gp130, leading to JAK1/STAT3 activation. Bile acids are steroid that modulates the expression/activity of membrane and nuclear receptors, including the Farnesoid-X-Receptor (FXR) and G Protein Bile Acid Activated Receptor (GPBAR1). Methods: Herein we have investigated whether ligands to FXR and GPBAR1 modulate LIF/LIFR pathway in PDAC cells and whether these receptors are expressed in human neoplastic tissues. Results: The transcriptome analysis of a cohort of PDCA patients revealed that expression of LIF and LIFR is increased in the neoplastic tissue in comparison to paired non-neoplastic tissues. By in vitro assay we found that both primary and secondary bile acids exert a weak antagonistic effect on LIF/LIFR signaling. In contrast, BAR502 a non-bile acid steroidal dual FXR and GPBAR1 ligand, potently inhibits binding of LIF to LIFR with an IC50 of 3.8 µM. Discussion: BAR502 reverses the pattern LIF-induced in a FXR and GPBAR1 independent manner, suggesting a potential role for BAR502 in the treatment of LIFR overexpressing-PDAC.

6.
Article in English | MEDLINE | ID: mdl-36411558

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic and relapsing disease caused by a dysregulated immune response to host intestinal microbiota that occurs in genetically predisposed individuals. IBD encompasses two major clinical entities: ulcerative colitis (UC), limited to the colonic mucosa, and Crohn's disease (CD), which might affect any segment of the gastrointestinal tract. Despite the prevalence of IBD increasing worldwide, therapy remains suboptimal, largely because of the variability of causative mechanisms, raising the need to develop individualized therapeutic approaches targeted to each individual patient. In this context, patients-derived intestinal organoids represent an effective tool for advancing our understanding of IBD's pathogenesis. Organoid 3D culture systems offer a unique model for dissecting epithelial mechanisms involved IBDs and testing individualized therapy, although the lack of a functional immune system and a microbiota, two driving components of the IBD pathogenesis, represent a major barrier to their exploitation in clinical medicine. In this review, we have examined how to improve the translational utility of intestinal organoids in IBD and how co-cultures of 3D or 2D organoids and immune cells and/or intestinal microbiota might help to overcome these limitations.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Humans , Intestines/pathology , Organoids/pathology
7.
Mol Cell Endocrinol ; 551: 111650, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35472625

ABSTRACT

The Farnesoid-x-receptor (FXR) is a bile acids sensor activated in humans by primary bile acids. FXR is mostly expressed in liver, intestine and adrenal glands but also by cells of innate immunity, including macrophages, liver resident macrophages, the Kupffer cells, natural killer cells and dendritic cells. In normal physiology and clinical disorders, cells of innate immunity mediate communications between liver, intestine and adipose tissues. In addition to FXR, the G protein coupled receptor (GPBAR1), that is mainly activated by secondary bile acids, whose expression largely overlaps FXR, modulates chemical communications from the intestinal microbiota and the host's immune system, integrating epithelial cells and immune cells in the entero-hepatic system, providing a mechanism for development of a tolerogenic state toward the intestinal microbiota. Disruption of FXR results in generalized inflammation and disrupted bile acids metabolism. While FXR agonism in preclinical models provides counter-regulatory signals that attenuate inflammation-driven immune dysfunction in a variety of liver and intestinal disease models, the clinical relevance of these mechanisms in the setting of FXR-related disorders remain poorly defined.


Subject(s)
Bile Acids and Salts , Receptors, Cytoplasmic and Nuclear , Bile Acids and Salts/metabolism , Humans , Inflammation/metabolism , Intestines , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism
8.
Biology (Basel) ; 11(4)2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35453818

ABSTRACT

The results of an ex-ante survey aiming to assess the impact of a fish farm in the Mar Grande of Taranto (southern Italy, Mediterranean Sea) on the surrounding environment are reported. There, the implementation of an innovative IMTA plant was planned, with the goals of environment bioremediation and commercially exploitable biomass production. Analyses were conducted in February and July 2018. Both seawater and sediments were sampled at the four corners of the fish farm to detect the existing biological and physico-chemical features. The investigation was performed to identify the best area of the farming plant for positioning the bioremediating system, but also to obtain a data baseline, to compare to the environmental status after the bioremediating action. Data were also analyzed by canonical analysis of principal coordinates (CAP). All the measurements, in particular, microbiology and macrobenthic community characterization using AZTI's Marine Biotic Index (AMBI) and the Multivariate-AMBI (M-AMBI) indices, suggest that the effect of fish farm waste was concentrated and limited to a small portion of the investigated area in relation to the direction of the main current. A site named A3, which was found to be the most impacted by the aquaculture activities, especially during the summer season, was chosen to place the bioremediation system.

9.
Expert Opin Drug Discov ; 16(10): 1193-1208, 2021 10.
Article in English | MEDLINE | ID: mdl-33849361

ABSTRACT

INTRODUCTION: The farnesoid-x-receptor (FXR) is a ubiquitously expressed nuclear receptor selectively activated by primary bile acids. AREA COVERED: FXR is a validated pharmacological target. Herein, the authors review preclinical and clinical data supporting the development of FXR agonists in the treatment of nonalcoholic fatty liver disease. EXPERT OPINION: Development of systemic FXR agonists to treat the metabolic liver disease has been proven challenging because the side effects associated with these agents including increased levels of cholesterol and LDL-c and reduced HDL-c raising concerns over their long-term cardiovascular safety. Additionally, pruritus has emerged as a common, although poorly explained, dose-related side effect with all FXR ligands, but is especially common with OCA. FXR agonists that are currently undergoing phase 2/3 trials are cilofexor, tropifexor, nidufexor and MET409. Some of these agents are currently being developed as combination therapies with other agents including cenicriviroc, a CCR2/CCR5 inhibitor, or firsocostat an acetyl CoA carboxylase inhibitor. Additional investigations are needed to evaluate the beneficial effects of combination of these agents with statins. It is expected that in the coming years, FXR agonists will be developed as a combination therapy to minimize side effects and increase likelihood of success by targeting different metabolic pathways.


Subject(s)
Azetidines , Non-alcoholic Fatty Liver Disease , Azetidines/therapeutic use , Chenodeoxycholic Acid/pharmacology , Chenodeoxycholic Acid/therapeutic use , Humans , Isonicotinic Acids/therapeutic use , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy
10.
Curr Opin Pharmacol ; 53: 45-54, 2020 08.
Article in English | MEDLINE | ID: mdl-32480317

ABSTRACT

Bile acids are produced in the liver by the cholesterol breakdown and further metabolized by the intestinal microbiota to generate a group of chemically heterogeneous steroids that bind and activate a family of cells surface and nuclear receptors, collectively known as the bile acid-activated receptors (BARs). The two best characterized members of this family are the farnesoid-x-receptor (FXR) and G protein Bile Acid Receptor (GPBAR1). Both receptors are expressed by cells of innate immunity including liver-resident and intestinal-resident macrophages and monocytes-derived macrophages. Because FXR and GPBAR1 knockout mice are biased toward a pro-inflammatory phenotype, it appears the both receptors might have a role in the development and maintenance of a tolerogenic phenotype. FXR and GPBAR1 ligands have been proven effective in the treatment in inflammatory and metabolic disorders and ligands for these receptors are currently under development for the treatment of non-alcoholic steato-hepatitis and diabetes.


Subject(s)
Macrophages/metabolism , Metabolic Diseases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Humans , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, G-Protein-Coupled/agonists
11.
Article in English | MEDLINE | ID: mdl-32371093

ABSTRACT

The farnesoid-X-receptor (FXR) is validated target in the cholestatic disorders treatment. Obeticholic acid (OCA), the first in class of FXR agonist approved for clinical use, causes side effects including acute liver decompensation when administered to cirrhotic patients with primary biliary cholangitis at higher than recommended doses. The V-Maf avian-musculoaponeurotic-fibrosarcoma-oncogene-homolog-G (Mafg) and nuclear factor-erythroid-2-related-factor-2 (Nrf2) mediates some of the downstream effects of FXR. In the present study we have investigated the role of FXR/MafG/NRF2 pathway in the development of liver toxicity caused by OCA in rodent models of cholestasis. Cholestasis was induced by bile duct ligation (BDL) or administration of α-naphtyl-isothiocyanate (ANIT) to male Wistar rats and FXR-/- and FXR+/+ mice. Treating BDL and ANIT rats with OCA exacerbated the severity of cholestasis, hepatocytes injury and severely downregulated the expression of basolateral transporters. In mice, genetic ablation FXR or its pharmacological inhibition by 3-(naphthalen-2-yl)-5-(piperidin-4-yl)-1,2,4-oxadiazole rescued from negative regulation of MRP4 and protected against liver injury caused by ANIT. By RNAseq analysis we found that FXR antagonism effectively reversed the transcription of over 2100 genes modulated by OCA/ANIT treatment, including Mafg and Nrf2 and their target genes Cyp7a1, Cyp8b1, Mat1a, Mat2a, Gss. Genetic and pharmacological Mafg inhibition by liver delivery of siRNA antisense or S-adenosylmethionine effectively rescued from damage caused by ANIT/OCA. In contrast, Nrf2 induction by sulforaphane was protective. CONCLUSIONS: Liver injury caused by FXR agonism in cholestasis is FXR-dependent and is reversed by FXR and Mafg antagonism or Nrf2 induction.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Cholestasis/metabolism , Liver Diseases/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Chenodeoxycholic Acid/pharmacology , Cholestasis/complications , Cholestasis/genetics , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver Diseases/etiology , Liver Diseases/genetics , MafG Transcription Factor/antagonists & inhibitors , MafG Transcription Factor/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics
12.
Nutrients ; 11(8)2019 Aug 07.
Article in English | MEDLINE | ID: mdl-31394746

ABSTRACT

Pelargonidins are anthocyanidins thought to be beneficial for the human health, although controversies exist over the doses needed and the unclear mechanism of action, along with poor systemic bioavailability. One putative target of pelargonidins is the aryl hydrocarbon receptor (AhR). A synthetic pelargonidin (Mt-P) was synthesized by the methylation of the pelargonidin (the natural compound indicated as P). Mt-P transactivated the AhR with an EC50 of 1.97 µM and was ~2-fold more potent than the natural compound. In vitro Mt-P attenuated pro-inflammatory activities of Raw264.7 macrophage cells in an AhR-dependent manner. In vivo, administration of the Mt-P in Balb/c mice resulted in a dose-dependent attenuation of signs and symptoms of colitis induced by TNBS. A dose of 5 mg/kg Mt-P, but not the natural compound P, reversed intestinal inflammation and increased expression of Tnf-α, Ifn-Æ´, and Il-6, while promoted the expansion of regulatory T cells and M2 macrophages. In C57BL/6J mice fed a high fat diet (HFD), Mt-P attenuated body weight gain, intestinal and liver inflammation, and ameliorated insulin sensitivity, while worsened liver steatosis by up-regulating the liver expression of Cd36 and Apo100b. These effects were abrogated by AhR gene ablation. Mt-P is a synthetic pelargonidin endowed with robust AhR agonist activity that exerts beneficial effects in murine models of inflammation and metabolic dysfunction.


Subject(s)
Anthocyanins/pharmacology , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Receptors, Aryl Hydrocarbon/physiology , Animals , Anthocyanins/chemistry , Caco-2 Cells , Colitis/chemically induced , Colitis/drug therapy , Fatty Liver/drug therapy , Hep G2 Cells , Humans , Macrophages/drug effects , Macrophages/physiology , Methylation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/genetics
13.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1422-1437, 2019 10.
Article in English | MEDLINE | ID: mdl-31325638

ABSTRACT

Obeticholic acid (OCA) is a farnesoid-X-receptor (FXR) ligand, shown effective in reducing steatosis and fibrosis in NASH patients. However, OCA causes major side effects including pruritus, while increases the risk for liver decompensation in cirrhotic patients. Ursodeoxycholic acid (UDCA), is a safe and unexpensive bile acid used in the treatment of liver disorders whose mechanism of action is poorly defined. Here we have compared the effects of OCA and UDCA in a mouse model of NASH. In mice exposed to a diet rich in fat/cholesterol and fructose (HFD-F), treatment with OCA or UDCA effectively prevented body weight gain, insulin resistance, as demonstrated by OGTT, and AST plasma levels. After 12 weeks HFD-F mice developed liver microvesicular steatosis, inflammation and mild fibrosis, increased expression of inflammatory (TNFα, IL6, F4/80) and fibrosis (αSma, Col1α1, Tgfß) markers, reduced liver expression of FXR, dysregulated liver FXR signaling and elevated levels of Tauro-α and ß-muricholic acid (T-α and ßMCA), two FXR antagonists in mice. Both compounds prevented these changes and improved liver histopathology. OCA reduced primary bile acid synthesis worsening the T-CA/T-ßMCA ratio. UDCA effectively transactivated GPBAR1 in vitro. By RNAseq analysis we found that among over 2400 genes modulated by the HFD-F, only 32 and 60 genes were modulated by OCA and UDCA, with only 3 genes (Dbp, Adh7, Osgin1) being modulated by both agents. Both agents partially prevented the intestinal dysbiosis. CONCLUSIONS: UDCA is a GPBAR1 ligand and exerts beneficial effects in a rodent model of NASH by activating non-overlapping pathway with OCA.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Dysbiosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/agonists , Ursodeoxycholic Acid/therapeutic use , Animals , Chenodeoxycholic Acid/therapeutic use , Diet/adverse effects , Dysbiosis/etiology , Dysbiosis/metabolism , Humans , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
14.
Front Immunol ; 10: 1157, 2019.
Article in English | MEDLINE | ID: mdl-31258525

ABSTRACT

The treatment of paroxysmal nocturnal hemoglobinuria has been revolutionized by the introduction of the anti-C5 agent eculizumab; however, eculizumab is not the cure for Paroxysmal nocturnal hemoglobinuria (PNH), and room for improvement remains. Indeed, the hematological benefit during eculizumab treatment for PNH is very heterogeneous among patients, and different response categories can be identified. Complete normalization of hemoglobin (complete and major hematological response), is seen in no more than one third of patients, while the remaining continue to experience some degree of anemia (good and partial hematological responses), in some cases requiring regular red blood cell transfusions (minor hematological response). Different factors contribute to residual anemia during eculizumab treatment: underlying bone marrow dysfunction, residual intravascular hemolysis and the emergence of C3-mediated extravascular hemolysis. These two latter pathogenic mechanisms are the target of novel strategies of anti-complement treatments, which can be split into terminal and proximal complement inhibitors. Many novel terminal complement inhibitors are now in clinical development: they all target C5 (as eculizumab), potentially paralleling the efficacy and safety profile of eculizumab. Possible advantages over eculizumab are long-lasting activity and subcutaneous self-administration. However, novel anti-C5 agents do not improve hematological response to eculizumab, even if some seem associated with a lower risk of breakthrough hemolysis caused by pharmacokinetic reasons (it remains unclear whether more effective inhibition of C5 is possible and clinically beneficial). Indeed, proximal inhibitors are designed to interfere with early phases of complement activation, eventually preventing C3-mediated extravascular hemolysis in addition to intravascular hemolysis. At the moment there are three strategies of proximal complement inhibition: anti-C3 agents, anti-factor D agents and anti-factor B agents. These agents are available either subcutaneously or orally, and have been investigated in monotherapy or in association with eculizumab in PNH patients. Preliminary data clearly demonstrate that proximal complement inhibition is pharmacologically feasible and apparently safe, and may drastically improve the hematological response to complement inhibition in PNH. Indeed, we envision a new scenario of therapeutic complement inhibition, where proximal inhibitors (either anti-C3, anti-FD or anti-FB) may prove effective for the treatment of PNH, either in monotherapy or in combination with anti-C5 agents, eventually leading to drastic improvement of hematological response.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Inactivating Agents/therapeutic use , Erythrocytes/immunology , Hemoglobinuria, Paroxysmal/drug therapy , Complement Activation/drug effects , Complement C3/antagonists & inhibitors , Complement C5/antagonists & inhibitors , Hemolysis/drug effects , Humans
15.
Nutrients ; 11(5)2019 May 21.
Article in English | MEDLINE | ID: mdl-31117231

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a progressive, chronic, liver disease whose prevalence is growing worldwide. Despite several agents being under development for treating NASH, there are no drugs currently approved. The Farnesoid-x-receptor (FXR) and the G-protein coupled bile acid receptor 1 (GPBAR1), two bile acid activated receptors, have been investigated for their potential in treating NASH. Here we report that BAR502, a steroidal dual ligand for FXR/GPBAR1, attenuates development of clinical and liver histopathology features of NASH in mice fed a high fat diet (HFD) and fructose (F). By RNAseq analysis of liver transcriptome we found that BAR502 restores FXR signaling in the liver of mice feed HFD-F, and negatively regulates a cluster of genes including Srebf1 (Srepb1c) and its target genes-fatty acid synthase (Fasn) and Cell death-inducing DFF45-like effector (CIDE) genes, Cidea and Cidec-involved in lipid droplets formation and triglycerides storage in hepatocytes. Additionally, BAR502 increased the intestinal expression of Fgf15 and Glp1 and energy expenditure by white adipose tissues. Finally, exposure to BAR502 reshaped the intestinal microbiota by increasing the amount of Bacteroidaceae. In conclusion, we have shown that dual FXR/GPBAR1 agonism might have utility in treatment of NASH.


Subject(s)
Cholanes/therapeutic use , Lipid Droplets/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Bile Acids and Salts/metabolism , Cholanes/pharmacology , Diet, High-Fat/adverse effects , Feces , Gastrointestinal Microbiome , Gene Expression Profiling , Gene Expression Regulation/drug effects , Ligands , Lipid Droplets/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, G-Protein-Coupled/genetics
16.
Nutrients ; 11(2)2019 Feb 02.
Article in English | MEDLINE | ID: mdl-30717413

ABSTRACT

A growing body of evidence suggests that probiotic functionality is not accurately predicted by their taxonomy. Here, we have set up a study to investigate the effectiveness of two probiotic formulations containing a blend of seven bacterial species in modulating intestinal inflammation in two rodent models of colitis, induced by treating mice with 2,4,6-Trinitrobenzenesulfonic acid (TNBS) or dextran sodium sulfate (DSS). Despite the taxonomy of the bacterial species in the two probiotic formulations being similar, only one preparation (Blend 2-Vivomixx) effectively attenuated the development of colitis in both models. In the TNBS model of colitis, Blend 2 reduced the expression of pro-inflammatory genes while increasing the production of anti-inflammatory cytokines, promoting the expansion M2 macrophages and the formation of IL-10-producing Treg cells in the colon's lamina propria. In the DSS model of colitis, disease attenuation and Treg formation was observed only in mice administered with Blend 2, and this effect was associated with intestinal microbiota remodeling and increased formation of lactate, butyrate, and propionate. None of these effects were observed in mice administered with Blend 1 (VSL#3). In summary, we have shown that two probiotic mixtures obtained by combining taxonomically similar species produced with different manufacturing methods exert divergent effects in mouse models of colitis.


Subject(s)
Bacteria , Colitis/microbiology , Colon/microbiology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Probiotics , Animals , Bacteria/genetics , Bacteria/growth & development , Bacteria/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/prevention & control , Colon/metabolism , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/microbiology , Inflammation/prevention & control , Inflammatory Bowel Diseases , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Male , Mice, Inbred BALB C , Probiotics/therapeutic use , Species Specificity , T-Lymphocytes, Regulatory/metabolism , Trinitrobenzenesulfonic Acid
17.
Bone Marrow Transplant ; 54(8): 1346-1353, 2019 08.
Article in English | MEDLINE | ID: mdl-30679824

ABSTRACT

Persistent cytopenia due to poor graft function (PoGF) is a relatively common complication which may affect up to 20% of patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT). Treatment options for PoGF remain limited, and reinfusion of additional HSC is often the only way to rescue hematopoiesis. Here we describe a retrospective single-center experience with the thrombopoietin-mimetic agent eltrombopag for the treatment of PoGF. Thirteen patients have received eltrombopag for either PoGF (n = 12) or primary graft failure (n = 1). In the 12 PoGF patients eltrombopag was started at the median time of 79 days after HSCT, due to persistent thrombocytopenia, with concomitant anemia and neutropenia in 7 and 3 patients, respectively. The treatment was started at the dose of 50 mg per day, and eventually increased up to 150 mg in case of lack of response. Hematological response was seen in 7 patients, with 6 complete responses. Hematological responses were seen both in patients with evidence of immune-mediated pathophysiology, and with possible infectious/iatrogenic causes. In responding patients, eltrombopag was discontinued in 6/7 patients without further relapse. These results suggest that eltrombopag is safe and possibly effective in the setting of the treatment of PoGF, and pave the way for future prospective studies.


Subject(s)
Benzoates/therapeutic use , Hydrazines/therapeutic use , Pyrazoles/therapeutic use , Thrombocytopenia/diet therapy , Adult , Aged , Benzoates/pharmacology , Female , Humans , Hydrazines/pharmacology , Male , Middle Aged , Pyrazoles/pharmacology , Retrospective Studies
19.
J Hematol Oncol ; 10(1): 126, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28629435

ABSTRACT

BACKGROUND: C5 blockade by eculizumab prevents complement-mediated intravascular hemolysis in paroxysmal nocturnal hemoglobinuria (PNH). However, C3-bound PNH red blood cells (RBCs), arising in almost all treated patients, may undergo extravascular hemolysis reducing clinical benefits. Despite the uniform deficiency of CD55 and of CD59, there are always two distinct populations of PNH RBCs, with (C3+) and without (C3-) C3 binding. METHODS: To investigate this paradox, the phenomenon has been modeled in vitro by incubating RBCs from eculizumab untreated PNH patients with compatible sera containing eculizumab, and by assessing the C3 binding after activation of complement alternative pathway. RESULTS: When RBCs from untreated patients were exposed in vitro to activated complement in the context of C5-blockade, there was the prompt appearance of a distinct C3+ PNH RBC population whose size increased with time and also with the rate of complement activation. Eventually, all PNH RBCs become C3+ to the same extent, without differences between old and young (reticulocytes) PNH RBCs. CONCLUSIONS: This study indicates that the distinct (C3+ and C3-) PNH RBC populations are not intrinsically different; rather, they result from a stochastic all-or-nothing phenomenon linked to the time-dependent cumulative probability of each individual PNH red cell to be exposed to levels of complement activation able to trigger C3 binding. These findings may envision novel approaches to reduce C3 opsonization and the subsequent extravascular hemolysis in PNH patients on eculizumab.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/drug effects , Complement C3/immunology , Erythrocytes/drug effects , Hemoglobinuria, Paroxysmal/drug therapy , Hemolysis/drug effects , CD59 Antigens/immunology , Complement C5/antagonists & inhibitors , Complement C5/immunology , Erythrocytes/immunology , Erythrocytes/pathology , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/pathology , Humans , Stochastic Processes
20.
Int J Surg Case Rep ; 32: 62-65, 2017.
Article in English | MEDLINE | ID: mdl-28249233

ABSTRACT

BACKGROUND: The management of a septic peritonitis open abdomen is a serious problem for clinicians. Open surgery is associated with several complications such as bleeding and perforation of the bowel. CASE PRESENTATION: The authors report a case of a 59-years-old female who underwent a sigmoid resection with an latero-terminal (L-T) anastomosis for the perforation of a diverticulum. After a few days the patients developed a new widespread peritonitis. At the emergency re-laparotomy, surgeons found dehiscence of the posterior wall of the anastomosis with fecal contamination. At admission in ICU (Intensive Care Unit) the patient had open abdomen with dehiscence of cutaneous and subcutaneous layers. CONCLUSION: Conservative therapy with antibiotic therapy and use of the Vacuum-Assisted Closure® (VAC) Therapy with a long term continuous saline infusion led to the resolution of the septic shock and to the wound healing.

SELECTION OF CITATIONS
SEARCH DETAIL
...