Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Can J Physiol Pharmacol ; 101(3): 147-159, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36744700

ABSTRACT

The antioxidant effect of caffeine, associated with its ability to upregulate the nuclear factor-E2-related factor-2 (Nrf2)-signaling pathway, was explored as a possible mechanism for the attenuation of liver damage. Nonalcoholic steatohepatitis (NASH) was induced in rats by the administration of a high-fat, high-sucrose, high-cholesterol diet (HFSCD) for 15 weeks. Liver damage was induced in rats by intraperitoneal administration of thioacetamide (TAA) for six weeks. Caffeine was administered orally at a daily dose of 50 mg/kg body weight during the period of NASH induction to evaluate its ability to prevent disease development. Meanwhile, rats received TAA for three weeks, after which 50 mg/kg caffeine was administered daily for three weeks with TAA to evaluate its capacity to interfere with the progression of hepatic injury. HFSCD administration induced hepatic steatosis, decreased Nrf2 levels, increased oxidative stress, induced the activation of nuclear factor-κB (NF-κB), and elevated proinflammatory cytokine levels, leading to hepatic damage. TAA administration produced similar effects, excluding steatosis. Caffeine increased Nrf2 levels; attenuated oxidative stress markers, including malondialdehyde and 4-hydroxynonenal; restored normal, reduced glutathione levels; and reduced NF-κB activation, inflammatory cytokine levels, and damage. Our findings suggest that caffeine may be useful in the treatment of human liver diseases.


Subject(s)
Antioxidants , Non-alcoholic Fatty Liver Disease , Humans , Rats , Male , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Thioacetamide/toxicity , Caffeine/pharmacology , Caffeine/therapeutic use , NF-kappa B/metabolism , NF-E2-Related Factor 2/metabolism , Liver , Oxidative Stress , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
2.
Ann Hepatol ; 28(1): 100780, 2023.
Article in English | MEDLINE | ID: mdl-36309184

ABSTRACT

INTRODUCTION AND OBJECTIVES: Administration of carbon tetrachloride (CCl4), along with an hepatopathogenic diet, is widely employed as a chemical inducer to replicate human nonalcoholic steatohepatitis (NASH) in rodents; however, the role of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome in this model remains unclear. We aimed to determine the relevance of NLRP3 inflammasome activation in the development of NASH induced by CCl4 along with an hepatopathogenic diet in male Wistar rats. MATERIALS AND METHODS: Animals were fed either a high fat, sucrose, and cholesterol diet (HFSCD) or a HFSCD plus intraperitoneal injections of low doses of CCl4 (400 mg/kg) once a week for 15 weeks. Liver steatosis, inflammation, fibrosis, and NLRP3 inflammasome activation were evaluated using biochemical, histological, ultrastructural, and immunofluorescence analyses, western blotting, and immunohistochemistry. RESULTS: Our experimental model reproduced several aspects of the human NASH pathophysiology. NLRP3 inflammasome activation was induced by the combined effect of HFSCD plus CCl4 and significantly increased levels of both proinflammatory and profibrogenic cytokines and collagen deposition in the liver; thus, NASH severity was higher in the HFSCD+CCl4 group than that in the HFSCD group, to which CCl4 was not administered. Hepatic stellate cells, the most profibrogenic cells, were activated by HFSCD plus CCl4, as indicated by elevated levels of α-smooth muscle actin. Thus, activation of the NLRP3 inflammasome, triggered by low doses of CCl4, exacerbates the severity of NASH. CONCLUSIONS: Our results indicate that NLRP3 inflammasome activation plays a key role and may be an important therapeutic target for NASH treatment.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Rats , Animals , Male , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Inflammasomes/adverse effects , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Wistar , Liver/pathology , Cholesterol , Diet, High-Fat/adverse effects , Mice, Inbred C57BL
3.
Ann Hepatol ; 27(2): 100671, 2022.
Article in English | MEDLINE | ID: mdl-35065262

ABSTRACT

INTRODUCTION AND OBJECTIVES: Caffeine consumption is associated with beneficial effects on hepatic disorders. The objectives of this study were to evaluate the antifibrotic effects of caffeine on experimental nonalcoholic steatohepatitis (NASH) induced with a high-fat, high-sucrose, high-cholesterol diet (HFSCD), as well as to evaluate the ability of caffeine to prevent the progression of experimental liver fibrosis induced by the administration of thioacetamide (TAA) in rats and explore the mechanisms of action. METHODS: NASH and fibrosis were induced in rats by the administration of an HFSCD for 15 weeks, and liver fibrosis was induced by intraperitoneal administration of 200 mg/kg TAA 3 times per week, for 6 weeks. Caffeine was administered at a dose of 50 mg/kg body weight. The effects of diet, TAA, and caffeine on fibrosis were evaluated by biochemical and histological examinations. The profibrotic pathways were analyzed by western blotting and immunohistochemistry. RESULTS: Rats exhibited liver fibrosis after HFSCD feeding and the administration of TAA. Caffeine could reduce the hepatic level of collagen and the fibrotic area in the liver. Caffeine prevented the progression of liver fibrosis by decreasing transforming growth factor-beta (TGF-ß), connective tissue growth factor (CTGF), and alpha-smooth muscle actin (α-SMA) expression and by inhibiting the activation of mitogen-activated protein kinases (MAPKs) and Smad3 phosphorylation. CONCLUSIONS: Caffeine attenuates NASH and the progression of liver fibrosis due to its antifibrotic effects and modulating the MAPK and TGF-ß pathways. Therefore, caffeine could be a suitable candidate for treating liver diseases associated with fibrosis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Thioacetamide , Animals , Caffeine/adverse effects , Caffeine/metabolism , Fibrosis , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/prevention & control , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Rats , Signal Transduction , Smad3 Protein/metabolism , Thioacetamide/adverse effects , Thioacetamide/metabolism , Transforming Growth Factor beta/metabolism
4.
Int J Mol Sci ; 23(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36077357

ABSTRACT

Caffeine elicits protective effects against liver diseases, such as NASH; however, its mechanism of action involving the pyrin domain-containing-3 (NLRP3) inflammasome signaling pathway remains to be elucidated. This study aimed to evaluate the effect of caffeine on the NLRP3 inflammasome signaling pathway in a rat model of NASH. NASH was induced by feeding rats a high-fat, -sucrose, and -cholesterol diet (HFSCD) for 15 weeks along with a weekly low dose (400 mg/kg, i.p.) of CCl4. Caffeine was administered at 50 mg/kg p.o. The effects of HFSCD+CCl4 and caffeine on the liver were evaluated using biochemical, ultrastructural, histological, and molecular biological approaches. The HFSCD+CCl4-treated rats showed fat accumulation in the liver, elevated levels of inflammatory mediators, NLRP3 inflammasome activation, antioxidant dysregulation, and liver fibrosis. Caffeine reduced necrosis, cholestasis, oxidative stress, and fibrosis. Caffeine exhibited anti-inflammatory effects by attenuating NLRP3 inflammasome activation. Moreover, caffeine prevented increases in toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) protein levels and mitigated the phosphorylation of mitogen-activated protein kinase (MAPK). Importantly, caffeine prevented the activation of hepatic stellate cells. This study is the first to report that caffeine ameliorates NASH by inhibiting NLRP3 inflammasome activation through the suppression of the TLR4/MAPK/NF-κB signaling pathway.


Subject(s)
NF-kappa B , Non-alcoholic Fatty Liver Disease , Animals , Caffeine/pharmacology , Caffeine/therapeutic use , Inflammasomes/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Signal Transduction , Toll-Like Receptor 4/metabolism
5.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31527129

ABSTRACT

Epidemiological studies suggest frequent association of enteropathogenic bacteria with Entamoeba histolytica during symptomatic infection. In this study, we sought to determine if the interaction with enteropathogenic (EPEC) or nonpathogenic Escherichia coli (strain DH5α) could modify the virulence of E. histolytica to cause disease in animal models of amebiasis. In vitro studies showed a 2-fold increase in CaCo2 monolayer destruction when E. histolytica interacted with EPEC but not with E. coli DH5α for 2.5 h. This was associated with increased E. histolytica proteolytic activity as revealed by zymogram analysis and degradation of the E. histolytica CP-A1/5 (EhCP-A1/5) peptide substrate Z-Arg-Arg-pNC and EhCP4 substrate Z-Val-Val-Arg-AMC. Additionally, E. histolytica-EPEC interaction increased EhCP-A1, -A2, -A4, and -A5, Hgl, Apa, and Cox-1 mRNA expression. Despite the marked upregulation of E. histolytica virulence factors, nonsignificant macroscopic differences in amebic liver abscess development were observed at early stages in hamsters inoculated with either E. histolytica-EPEC or E. histolytica-E. coli DH5α. Histopathology of livers of E. histolytica-EPEC-inoculated animals revealed foci of acute inflammation 3 h postinoculation that progressively increased, producing large inflammatory reactions, ischemia, and necrosis with high expression of il-1ß, ifn-γ, and tnf-α proinflammatory cytokine genes compared with that in livers of E. histolytica-E. coli DH5α-inoculated animals. In closed colonic loops from mice, intense inflammation was observed with E. histolytica-EPEC manifested by downregulation of Math1 mRNA with a corresponding increase in the expression of Muc2 mucin and proinflammatory cytokine genes il-6, il-12, and mcp-1 These results demonstrate that E. histolytica/EPEC interaction enhanced the expression and production of key molecules associated with E. histolytica virulence, critical in pathogenesis and progression of disease.


Subject(s)
Entamoeba histolytica/pathogenicity , Entamoebiasis/pathology , Enteropathogenic Escherichia coli/physiology , Host Microbial Interactions/physiology , Animals , Caco-2 Cells , Cell Line , Cricetinae , Cysteine Proteases/metabolism , Cytokines/metabolism , Entamoeba histolytica/microbiology , HT29 Cells , Humans , Inflammation , Mesocricetus , Mice , Mice, Inbred C57BL , Mucin-2/metabolism , Virulence Factors/biosynthesis
6.
Hepatol Res ; 49(2): 212-223, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30338893

ABSTRACT

AIM: The aims of the present study were to investigate the capacity of stevia leaves to prevent experimental cirrhosis induced by chronic administration of carbon tetrachloride (CCl4 ) in rats and to explore the action mechanism involved. METHODS: Liver cirrhosis was established by CCl4 treatment (400 mg/kg i.p. three times a week for 12 weeks); stevia powder was administered (100 mg/kg by gavage daily) during the CCl4 treatment. Serum markers of liver damage and hydroxyproline were evaluated and histopathological analyses were carried out. The profibrotic pathways were analyzed by western blot and immunohistochemistry. RESULTS: We found for the first time that stevia cotreatment prevented the elevation of serum markers of necrosis and cholestasis and the occurrence of liver fibrosis. It is worth noting that stevia downregulated several profibrogenic pathways, including the reduction of hepatic myofibroblasts and decreased matrix metalloproteinase (MMP)2 and MMP13 expression, thereby blocking the liberation of transforming growth factor-ß from the extracellular matrix. Notably, stevia reduced the phosphorylation of pSmad3L, the most profibrogenic and mitogenic Smad, by inhibiting the activation of c-Jun N-terminal kinase and extracellular signal-regulated kinase. Interestingly, Smad7, an important antifibrotic molecule, was upregulated by stevia treatment in cirrhotic rats. These multitarget mechanisms led to the prevention of experimental cirrhosis. CONCLUSIONS: Because stevia possesses a reasonable safety profile, our results indicate that it could be useful in the clinical setting to treat chronic liver diseases.

7.
Ann Hepatol ; 18(3): 472-479, 2019.
Article in English | MEDLINE | ID: mdl-31053541

ABSTRACT

INTRODUCTION AND AIM: Stevia has exhibited antioxidant, antihyperglycemic, antihypertensive and anti-inflammatory properties in several in vivo and in vitro models. The objective of this study was to investigate the ability of an aqueous extract of stevia (AES) to prevent experimental cirrhosis in rats and to explore its mechanism of action. MATERIALS AND METHODS: Liver cirrhosis was induced by administering carbon tetrachloride (CCl4) (400mg/kg by i.p. injection 3 times a week for 12 weeks); AES was administered (100mg/kg by gavage daily) during the CCl4 treatment. Fibrosis was evaluated with histological, biochemical and molecular approaches, and liver damage was assessed with standardized procedures. The profibrotic pathways were analyzed by western blotting, qRT-PCR and immunohistochemistry. RESULTS AND CONCLUSIONS: Chronic CCl4 administration increased nuclear factor kappa B (NF-κB) and proinflammatory cytokine production as well as oxidative parameters such as lipid peroxidation and 4-hydroxynonenal levels, whereas GSH and nuclear factor-E2-related factor 2 (Nrf2) levels were decreased. CCl4 induced profibrogenic mediator expression, hepatic stellate cell (HSC) activation and, consequently, extracellular matrix production. AES exhibited antioxidant, anti-inflammatory and antifibrotic properties, probably because of its capacity to induce Nrf2 expression, reduce NF-κB expression and block several profibrogenic signaling pathways, subsequently inhibiting HSC activation and preventing fibrosis induced by chronic CCl4 administration.


Subject(s)
Liver Cirrhosis, Experimental/prevention & control , Liver/pathology , Oxidative Stress , Plant Extracts/pharmacology , Stevia , Animals , Humans , Liver/drug effects , Liver/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Rats , Rats, Wistar , Sweetening Agents/pharmacology
8.
Phytother Res ; 32(12): 2568-2576, 2018 12.
Article in English | MEDLINE | ID: mdl-30251285

ABSTRACT

Stevia has been shown to prevent oxidative stress and inflammation in carbon tetrachloride­induced cirrhosis models. This study aimed to investigate the ability of an aqueous extract of stevia (AES) to prevent thioacetamide (TAA)­induced cirrhosis in rats and to explore its mechanism of action. Liver cirrhosis was established by administering TAA (200 mg/kg by i.p. injections three times a week for 10 weeks); AES was administered (100 mg/kg by gavage daily) during the TAA treatment. Liver damage and fibrosis were evaluated, and the profibrotic pathways were analyzed by western blotting and immunohistochemistry. TAA increased nuclear factor kappa B (NF­κB) and pro­inflammatory cytokine production, as well as the malondialdehyde and 4­hydroxynonenal levels, whereas the glutathione/glutathione disulfide and nuclear factor­E2­related factor 2 (Nrf2) levels were decreased. Moreover, TAA increased collagen production, hepatic stellate cell (HSC) activation, and expression of profibrogenic mediators. TAA­treated rats that had been exposed to Mn2+ exhibited altered striatal dopamine turnover, indicating hepatic encephalopathy. AES partially or completely prevented all of these effects. AES showed antioxidant, anti­inflammatory, and antifibrotic properties, probably because of its capacity to induce Nrf2 expression, reduce NF­κB expression, and block several profibrogenic signaling pathways, subsequently inhibiting HSC activation and preventing fibrosis and dopamine turnover.


Subject(s)
Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/prevention & control , NF-E2-Related Factor 2/physiology , NF-kappa B/physiology , Plant Extracts/therapeutic use , Smad7 Protein/physiology , Stevia , Transforming Growth Factor beta/physiology , Animals , Hepatic Stellate Cells/physiology , Liver Cirrhosis, Experimental/chemically induced , Male , Rats , Rats, Wistar , Signal Transduction/drug effects , Thioacetamide
9.
Antimicrob Agents Chemother ; 58(3): 1523-8, 2014.
Article in English | MEDLINE | ID: mdl-24366747

ABSTRACT

Painful blinding keratitis and fatal granulomatous amebic encephalitis are caused by the free-living amebae Acanthamoeba spp. Several prescription eye medications are used to treat Acanthamoeba keratitis, but the infection can be difficult to control because of recurrence of infection. For the treatment of encephalitis, no single drug was found useful, and in spite of the use of a combination of multiple drugs, the mortality rate remains high. Therefore, efficient, novel drugs are urgently needed for the treatment of amebic keratitis and granulomatous amebic encephalitis. In this study, we identified corifungin, a water-soluble polyene macrolide, as amebicidal. In vitro, it was effective against both the trophozoites and the cysts. Transmission electron microscopy of Acanthamoeba castellanii incubated with corifungin showed the presence of swollen mitochondria, electron-dense granules, degeneration of cytoplasm architecture, and loss of nuclear chromatin structure. These changes were followed by lysis of amebae. Corifungin also induced the encystment process of A. castellanii. There were alterations in the cyst cell wall followed by lysis of the cysts. Corifungin is a promising therapeutic option for keratitis and granulomatous amebic encephalitis.


Subject(s)
Acanthamoeba castellanii/drug effects , Amebicides/pharmacology , Aminoglycosides/pharmacology , Macrolides/pharmacology , Acanthamoeba Keratitis/drug therapy , Acanthamoeba castellanii/ultrastructure , Cell Wall/drug effects , Encephalitis/drug therapy , Encephalitis/parasitology , In Vitro Techniques , Microscopy, Electron, Transmission , Mitochondria/drug effects , Trophozoites/drug effects
10.
Microbiology (Reading) ; 159(Pt 2): 392-401, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23258265

ABSTRACT

Naegleria fowleri is the aetiological agent of primary amoebic meningoencephalitis. This parasite invades its host by penetrating the olfactory mucosa. However, the mechanism of epithelium penetration is not well understood. In the present study, we evaluated the effect of N. fowleri trophozoites and the non-pathogenic Naegleria gruberi on Madin-Darby canine kidney (MDCK) tight junction proteins, including claudin-1, occludin and ZO-1, as well as on the actin cytoskeleton. Trophozoites from each of the free-living amoeba species were co-cultured with MDCK cells in a 1 : 1 ratio for 1, 3, 6 or 10 h. Light microscopy revealed that N. fowleri caused morphological changes as early as 3 h post-infection in an epithelial MDCK monolayer. Confocal microscopy analysis revealed that after 10 h of co-culture, N. fowleri trophozoites induced epithelial cell damage, which was characterized by changes in the actin apical ring and disruption of the ZO-1 and claudin-1 proteins but not occludin. Western blot assays revealed gradual degradation of ZO-1 and claudin-1 as early as 3 h post-infection. Likewise, there was a drop in transepithelial electrical resistance that resulted in increased epithelial permeability and facilitated the invasion of N. fowleri trophozoites by a paracellular route. In contrast, N. gruberi did not induce alterations in MDCK cells even at 10 h post-infection. Based on these results, we suggest that N. fowleri trophozoites disrupt epithelial monolayers, which could enable their penetration of the olfactory epithelium and subsequent invasion of the central nervous system.


Subject(s)
Naegleria fowleri/pathogenicity , Tight Junction Proteins/metabolism , Tight Junctions/microbiology , Actins/metabolism , Animals , Blotting, Western , Coculture Techniques , Dogs , Madin Darby Canine Kidney Cells , Microscopy
11.
Xenobiotica ; 43(2): 161-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22834477

ABSTRACT

Cytochrome P4502E1 (CYP2E1) is involved in the biotransformation of several low molecular weight chemicals and plays an important role in the metabolic activation of carcinogens and hepatotoxins such as CCl(4). Induction of CYP2E1 is exerted mainly at posttranscriptional levels through mRNA and protein stabilization, and there is little evidence of xenobiotic induction at the transcriptional level. Previously, we reported microarray analysis data suggesting a decrease in Cyp2e1 gene expression on Ahr-null livers when compared to wild-type mouse livers. The goal of the present study was to determine whether 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) increased mouse CYP2E1 levels in an AhR-dependent manner and the impact on CCl(4)-induced hepatotoxicity. TCDD treatment induced CYP2E1 mRNA and protein levels in mouse liver, and this effect was aryl hydrocarbon receptor (AhR)-dependent. Moreover, TCDD pre-treatment increased the CCl(4)-induced alanine aminotransferase (ALT) activity, the extent of CCl(4)-induced necrosis, and the number of sinusoidal cells in wild-type animals, while this potentiating effect was not observed in Ahr-null mice. In conclusion, this study revealed that TCDD, probably in an AhR-dependent manner, exacerbated CCl(4)-induced hepatotoxicity through induction of CYP2E1.


Subject(s)
Carbon Tetrachloride/toxicity , Cytochrome P-450 CYP2E1/metabolism , Environmental Pollutants/toxicity , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Alanine Transaminase/metabolism , Animals , Liver/drug effects , Liver/enzymology , Male , Mice
12.
Antimicrob Agents Chemother ; 56(11): 5450-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22869574

ABSTRACT

Primary amebic meningoencephalitis (PAM) is a rapidly fatal infection caused by the free-living ameba Naegleria fowleri. The drug of choice in treating PAM is the antifungal antibiotic amphotericin B, but its use is associated with severe adverse effects. Moreover, few patients treated with amphotericin B have survived PAM. Therefore, fast-acting and efficient drugs are urgently needed for the treatment of PAM. To facilitate drug screening for this pathogen, an automated, high-throughput screening methodology was developed and validated for the closely related species Naegleria gruberi. Five kinase inhibitors and an NF-kappaB inhibitor were hits identified in primary screens of three compound libraries. Most importantly for a preclinical drug discovery pipeline, we identified corifungin, a water-soluble polyene macrolide with a higher activity against Naegleria than that of amphotericin B. Transmission electron microscopy of N. fowleri trophozoites incubated with different concentrations of corifungin showed disruption of cytoplasmic and plasma membranes and alterations in mitochondria, followed by complete lysis of amebae. In vivo efficacy of corifungin in a mouse model of PAM was confirmed by an absence of detectable amebae in the brain and 100% survival of mice for 17 days postinfection for a single daily intraperitoneal dose of 9 mg/kg of body weight given for 10 days. The same dose of amphotericin B did not reduce ameba growth, and mouse survival was compromised. Based on these results, the U.S. FDA has approved orphan drug status for corifungin for the treatment of PAM.


Subject(s)
Amebiasis/drug therapy , Aminoglycosides/pharmacology , Antiprotozoal Agents/pharmacology , Central Nervous System Protozoal Infections/drug therapy , Macrolides/pharmacology , Naegleria fowleri/drug effects , Naegleria/drug effects , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Trophozoites/drug effects , Amebiasis/mortality , Amebiasis/parasitology , Aminoglycosides/chemistry , Amphotericin B/chemistry , Amphotericin B/pharmacology , Animals , Antiprotozoal Agents/chemistry , Brain/drug effects , Brain/parasitology , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Central Nervous System Protozoal Infections/mortality , Central Nervous System Protozoal Infections/parasitology , Drug Administration Schedule , High-Throughput Screening Assays , Humans , Injections, Intraperitoneal , Macrolides/chemistry , Mice , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/ultrastructure , NF-kappa B/antagonists & inhibitors , Naegleria/growth & development , Naegleria/ultrastructure , Naegleria fowleri/growth & development , Naegleria fowleri/ultrastructure , Protein Kinase Inhibitors/chemistry , Protein Kinases/metabolism , Small Molecule Libraries/chemistry , Survival Rate , Trophozoites/growth & development , Trophozoites/ultrastructure
13.
Front Cell Infect Microbiol ; 12: 885191, 2022.
Article in English | MEDLINE | ID: mdl-35706909

ABSTRACT

Typical enteroaggregative Escherichia coli (tEAEC) is a diarrheagenic E. coli pathotype associated with pediatric and traveler's diarrhea. Even without diarrhea, EAEC infections in children also lead to increased gut inflammation and growth shortfalls. EAEC strain's defining phenotype is the aggregative adherence pattern on epithelial cells attributable to the aggregative adherence fimbriae (AAF). EAEC only causes diarrhea in humans; therefore, not much is known of the exact intestinal region of infection and damage or its interactions with intestinal enterocytes in vivo and in situ. This study aimed to develop a new tEAEC mouse model of infection, characterize the microbiota of infected mice, and evaluate in situ the expression of host adherence and surface molecules triggering EAEC infection and the role of the EAEC AAF-II in adherence. Six-week-old C57BL/6 mice, without previous antibiotic treatment, were orally challenged with EAEC 042 strain or EAEC 042 AAF-II mutant (ΔAAF/II) strain, or DAEC-MXR strain (diffusely adherent E. coli clinical isolate), and with saline solution (control group). Paraffin sections of the colon and ileum were stained with H&E and periodic acid-Schiff. ZO-1, ß-catenin, MUC1, and bacteria were analyzed by immunofluorescence. EAEC-infected mice, in comparison with DAEC-MXR-infected and control mice, significantly lost weight during the first 3 days. After 7 days post-infection, mucus production was increased in the colon and ileum, ZO-1 localization remained unaltered, and morphological alterations were more pronounced in the ileum since increased expression and apical localization of ß-catenin in ileal enterocytes were observed. EAEC-infected mice developed dysbiosis 21 days post-infection. At 4 days post-infection, EAEC strain 042 formed a biofilm on ileal villi and increased the expression and apical localization of ß-catenin in ileal enterocytes; these effects were not seen in animals infected with the 042 ΔAAF/II strain. At 3 days post-infection, MUC1 expression on ileal enterocytes was mainly detectable among infected mice and colocalized with 042 strains on the enterocyte surface. We developed a novel mouse model of EAEC infection, which mimics human infection, not an illness, revealing that EAEC 042 exerts its pathogenic effects in the mouse ileum and causes dysbiosis. This model is a unique tool to unveil early molecular mechanisms of EAEC infection in vivo and in situ.


Subject(s)
Escherichia coli Infections , Ileum , Microbiota , Mucin-1 , beta Catenin , Adhesins, Escherichia coli/genetics , Animals , Bacterial Adhesion/genetics , Diarrhea/microbiology , Disease Models, Animal , Dysbiosis , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Humans , Mice , Mice, Inbred C57BL , Mucin-1/genetics , Mucus/metabolism , Travel , beta Catenin/genetics
14.
Parasitol Res ; 108(2): 439-49, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20922421

ABSTRACT

Entamoeba histolytica calreticulin (EhCRT) is remarkably immunogenic in humans (90-100% of invasive amoebiasis patients). Nevertheless, the study of calreticulin in this protozoan is still in its early stages. The exact location, biological functions, and its role in pathogenesis are yet to be fully understood. The aim of the present work is to determine the location of EhCRT in virulent trophozoites in vivo and the expression of the Ehcrt gene during the development of experimentally induced amoebic liver abscesses (ALA) in hamsters. Antibodies against recombinant EhCRT were used for the immunolocalization of EhCRT in trophozoites through confocal microscopy; immunohistochemical assays were also performed on tissue sections of ALAs at different times after intrahepatic inoculation. The expression of the Ehcrt gene during the development of ALA was estimated through both in situ RT-PCR and real-time RT-PCR. Confocal assays of virulent trophozoites showed a distribution of EhCRT in the cytoplasmic vesicles of different sizes. Apparently, EhCRT is not exported into the hepatic tissue. Real-time RT-PCR demonstrated an over-expression of the Ehcrt gene at 30 min after trophozoite inoculation, reaching a peak at 1-2 h; thereafter, the expression fell sharply to its original levels. These results demonstrate for the first time in an in vivo model of ALA, the expression of Ehcrt gene in E. histolytica trophozoites and add evidence that support CRT as a resident protein of the ER in E. histolytica species. The in vivo experiments suggest that CRT may play an important role during the early stages of the host-parasite relationship, when the parasite is adapting to a new environment, although the protein seems to be constitutively synthesized. Moreover, trophozoites apparently do not export EhCRT into the hepatic tissue in ALA.


Subject(s)
Calreticulin/metabolism , Entamoeba histolytica/metabolism , Liver Abscess, Amebic/metabolism , Protozoan Proteins/metabolism , Trophozoites/metabolism , Animals , Blotting, Western , Calreticulin/genetics , Calreticulin/immunology , Cricetinae , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Entamoeba histolytica/genetics , Entamoeba histolytica/immunology , Gene Amplification , Gene Expression , Host-Parasite Interactions , Liver/metabolism , Liver/parasitology , Liver/pathology , Liver Abscess, Amebic/parasitology , Liver Abscess, Amebic/pathology , Mesocricetus , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Recombinant Proteins
15.
J Immunol Res ; 2021: 6624246, 2021.
Article in English | MEDLINE | ID: mdl-33928168

ABSTRACT

Leishmaniasis is a disease caused by an intracellular protozoan parasite of the genus Leishmania. Current treatments for leishmaniasis are long, toxic, and expensive and are not available in some endemic regions. Attempts to develop an effective vaccine are feasible, but no vaccine is in active clinical use. In this study, the LmxMBA gene of Leishmania mexicana was selected as a possible vaccine candidate using the reverse vaccinology approach, and the prophylactic effect generated by DNA vaccination with this gene in a murine model of cutaneous leishmaniasis was evaluated. The results showed that prophylactic vaccination with pVAX1::LmxMBA significantly reduced the size of the lesion and the parasitic load on the footpad, compared to the control groups. At a histological level, a smaller number of parasites were evident in the dermis, as well as the absence of connective tissue damage. Mice immunized with plasmid pVAX1::LmxMBA induced immunity characterized by an increase in the IgG2a/IgG1 > 1 ratio and a higher rate of lymphocyte proliferation. In this study, immunization with the plasmid promoted an improvement in the macroscopic and microscopic clinical manifestations of the experimental infection by L. mexicana, with a T helper 1 response characterized by an IgG2a/IgG1 > 1 ratio and high lymphoproliferative response. These findings support immunization with the plasmid pVAX1::LmxMBA as a preventive strategy against cutaneous infection of L. mexicana.


Subject(s)
Acid Phosphatase/genetics , Leishmania mexicana/physiology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Cutaneous/immunology , Protozoan Proteins/genetics , Skin/pathology , Th1 Cells/immunology , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Female , Humans , Immunoglobulin G/blood , Leishmaniasis, Cutaneous/prevention & control , Mice , Mice, Inbred BALB C , Parasite Load , Vaccination , Vaccines, DNA
16.
Can J Microbiol ; 56(12): 987-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21164568

ABSTRACT

Trophozoites of Entamoeba histolytica HM-1:IMSS become less virulent after long-term maintenance in axenic cultures. The factors responsible for the loss of virulence during in vitro cultivation remain unclear. However, it is known that in vitro cultivation of amoeba in culture medium supplemented with cholesterol restores their virulence. In this study, we analyzed the effect of adding phosphatidylcholine-cholesterol (PC-Chol) liposomes to the culture medium and evaluated the effect of this lipid on various biochemical and biological functions of E. histolytica HM-1:IMSS in terms of its virulence. The addition of PC-Chol liposomes to the culture medium maintained the virulence of these parasites against hamster liver at the same level as the original virulent E. histolytica strain, even though these amoebae were maintained without passage through hamster liver for 18 months. The trophozoites also showed increased endocytosis, erythrophagocytosis, and carbohydrate residue expression on the amoebic surface. Protease activities were also modified by the presence of cholesterol in the culture medium. These findings indicate the capacity of cholesterol to preserve amoeba virulence and provide an alternative method for the maintenance of virulent E. histolytica trophozoites without the need for in vivo procedures.


Subject(s)
Cholesterol/pharmacology , Entamoeba histolytica/drug effects , Entamoeba histolytica/pathogenicity , Liver Abscess, Amebic/parasitology , Phosphatidylcholines/pharmacology , Animals , Cholesterol/analysis , Concanavalin A/analysis , Cricetinae , Culture Media/chemistry , Endocytosis/drug effects , Entamoeba histolytica/enzymology , Entamoeba histolytica/growth & development , Erythrocytes/drug effects , Liposomes/pharmacology , Male , Peptide Hydrolases/metabolism , Phagocytosis/drug effects , Trophozoites/drug effects , Trophozoites/enzymology , Trophozoites/growth & development , Virulence/drug effects , Virulence Factors/metabolism
17.
Oxid Med Cell Longev ; 2018: 3823426, 2018.
Article in English | MEDLINE | ID: mdl-29849889

ABSTRACT

The effect of stevia on liver cirrhosis has not been previously investigated. In the present study, the antioxidant and anti-inflammatory properties of stevia leaves were studied in male Wistar rats with carbon tetrachloride- (CCl4-) induced acute and chronic liver damage. Acute and chronic liver damage induced oxidative stress, necrosis, and cholestasis, which were significantly ameliorated by stevia. Chronic CCl4 treatment resulted in liver cirrhosis, as evidenced by nodules of hepatocytes surrounded by thick bands of collagen and distortion of the hepatic architecture, and stevia significantly prevented these alterations. Subsequently, the underlying mechanism of action of the plant was analyzed. Our study for the first time shows that stevia upregulated Nrf2, thereby counteracting oxidative stress, and prevented necrosis and cholestasis through modulation of the main proinflammatory cytokines via NF-κB inhibition. These multitarget mechanisms led to the prevention of experimental cirrhosis. Given the reasonable safety profile of stevia, our results indicated that it may be useful for the clinical treatment of acute and chronic liver diseases.


Subject(s)
Carbon Tetrachloride/adverse effects , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Chemical and Drug Induced Liver Injury/drug therapy , Liver Cirrhosis/drug therapy , Liver/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Stevia/chemistry , Animals , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury, Chronic/pathology , Humans , Liver Cirrhosis/pathology , Male , Pilot Projects , Rats , Rats, Wistar , Up-Regulation
18.
Rev Invest Clin ; 59(5): 342-7, 2007.
Article in English | MEDLINE | ID: mdl-18268889

ABSTRACT

The protozoon Naegleria fowleri (N. fowleri) is a free-living amoeba that produces primary amoebic meningoencephalitis (PAM), which is an acute and frequently fatal infection of the central nervous system. We characterized the strains of N. fowleri isolated from the cerebrospinal fluid (CSF) of two cases presented in northwestern Mexico. The strains were isolated and cultured in 2% bactocasitone medium. Enflagellation assays, ultrastructural analysis, protein and protease electrophoresis patterns, and PCR were performed as confirmatory tests. Virulence tests were done using in Balb/c mice. Light microscopy analysis of brain tissue showed amoebae with abundant inflammatory reaction and extensive necrotic and hemorrhagic areas. The enflagellation assay was positive and the electron microscopy showed trophozoites with morphologic features typical of the genus. Protein and protease profiles of the isolated strains were identical to the reference strain. Finally, a 1500-bp PCR product was found in all three strains. Based on all the analyses performed, we concluded that the etiologic agent of both PAM cases was N. fowleri. The need for better epidemiological information and educational programs about basic clinical and pathological aspects of free-living amoebae provided by the health authorities are emphasized.


Subject(s)
Amebiasis/parasitology , Central Nervous System Protozoal Infections/parasitology , Naegleria fowleri/isolation & purification , Adult , Animals , Child , Humans , Male
19.
Biomed Res Int ; 2015: 679850, 2015.
Article in English | MEDLINE | ID: mdl-26064940

ABSTRACT

Individuals with X-HIGM syndrome fail to express functional CD40 ligand; consequently they cannot mount effective protective antibody responses against pathogenic bacteria. We evaluated, compared, and characterized the humoral immune response of wild type (WT) and C57-CD40L deficient (C57-CD40L(-/-)) mice infected with Citrobacter rodentium. Basal serum isotype levels were similar for IgM and IgG3 among mice, while total IgG and IgG2b concentrations were significantly lower in C57-CD40L(-/-) mice compared with WT. Essentially IgG1 and IgG2c levels were detectable only in WT mice. C57-CD40L(-/-) animals, orally inoculated with 2 × 10(9) CFU, presented several clinical manifestations since the second week of infection and eventually died. In contrast at this time point no clinical manifestations were observed among C57-CD40L(-/-) mice infected with 1 × 10(7) CFU. Infection was subclinical in WT mice inoculated with either bacterial dose. The serum samples from infected mice (1 × 10(7) CFU), collected at day 14 after infection, had similar C. rodentium-specific IgM titres. Although C57-CD40L(-/-) animals had lower IgG and IgG2b titres than WT mice, C57-CD40L(-/-) mice sera displayed complement-mediated bactericidal activity against C. rodentium. C. rodentium-infected C57-CD40L(-/-) mice are capable of producing antibodies that are protective. C57-CD40L(-/-) mouse is a useful surrogate model of X-HIGM syndrome for studying immune responses elicited against pathogens.


Subject(s)
CD40 Antigens/biosynthesis , Hyper-IgM Immunodeficiency Syndrome, Type 1/immunology , Immunity, Humoral/genetics , Immunoglobulin M/immunology , Animals , CD40 Antigens/immunology , Citrobacter rodentium/pathogenicity , Disease Models, Animal , Gene Expression Regulation , Humans , Hyper-IgM Immunodeficiency Syndrome, Type 1/genetics , Hyper-IgM Immunodeficiency Syndrome, Type 1/pathology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Ligands , Mice , Mice, Knockout
20.
Biomed Res Int ; 2014: 127453, 2014.
Article in English | MEDLINE | ID: mdl-24860808

ABSTRACT

The role of calreticulin (CRT) in host-parasite interactions has recently become an important area of research. Information about the functions of calreticulin and its relevance to the physiology of Entamoeba parasites is limited. The present work demonstrates that CRT of both pathogenic E. histolytica and nonpathogenic E. dispar species specifically interacted with human C1q inhibiting the activation of the classical complement pathway. Using recombinant EhCRT protein, we demonstrate that CRT interaction site and human C1q is located at the N-terminal region of EhCRT. The immunofluorescence and confocal microscopy experiments show that CRT and human C1q colocalize in the cytoplasmic vesicles and near to the surface membrane of previously permeabilized trophozoites or are incubated with normal human serum which is known to destroy trophozoites. In the presence of peripheral mononuclear blood cells, the distribution of EhCRT and C1q is clearly over the surface membrane of trophozoites. Nevertheless, the level of expression of CRT in situ in lesions of amoebic liver abscess (ALA) in the hamster model is different in both Entamoeba species; this molecule is expressed in higher levels in E. histolytica than in E. dispar. This result suggests that EhCRT may modulate some functions during the early moments of the host-parasite relationship.


Subject(s)
Calreticulin/immunology , Complement Pathway, Classical/immunology , Entamoeba histolytica/immunology , Entamoeba histolytica/pathogenicity , Liver Abscess, Amebic/immunology , Liver Abscess, Amebic/parasitology , Gene Expression Regulation/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL