Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pflugers Arch ; 476(5): 755-767, 2024 May.
Article in English | MEDLINE | ID: mdl-38305876

ABSTRACT

It has been suggested that the novel selective phosphodiesterase 9 (PDE9) inhibitor may improve cardiac and renal function by blocking 3',5'-cyclic guanosine monophosphate (cGMP) degradation. 5/6 nephrectomized (5/6Nx) rats were used to investigate the effects of the PDE9 inhibitor (BAY 73-6691) on the heart and kidney. Two doses of BAY 73-6691 (1 mg/kg/day and 5 mg/kg/day) were given for 95 days. The 5/6Nx rats developed albuminuria, a decrease in serum creatinine clearance (Ccr), and elevated serum troponin T levels. Echocardiographic data showed that 5/6 nephrectomy resulted in increased fractional shortening (FS), stroke volume (SV), and left ventricular ejection fraction (EF). However, 95 days of PDE9 inhibitor treatment did not improve any cardiac and renal functional parameter. Histopathologically, 5/6 nephrectomy resulted in severe kidney and heart damage, such as renal interstitial fibrosis, glomerulosclerosis, and enlarged cardiomyocytes. Telmisartan attenuated renal interstitial fibrosis and glomerulosclerosis as well as improved cardiomyocyte size. However, except for cardiomyocyte size and renal perivascular fibrosis, BAY 73-6691 had no effect on other cardiac and renal histologic parameters. Pathway enrichment analysis using RNA sequencing data of kidney and heart tissue identified chronic kidney disease pathways, such as phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) signaling pathway, complement and coagulation cascades, and nuclear factor kappa B (NF-κB) signaling pathway. PDE9i did not affect any of these disease-related pathways. Two dosages of the PDE9 inhibitor BAY 73-6691 known to be effective in other rat models have only limited cardio-renal protective effects in 5/6 nephrectomized rats.


Subject(s)
Heart , Kidney , Nephrectomy , Animals , Male , Rats , Heart/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Myocardium/metabolism , Myocardium/pathology , Nephrectomy/methods
2.
Am J Physiol Cell Physiol ; 324(4): C951-C962, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36779666

ABSTRACT

The mechanisms of nephroprotection in nondiabetic chronic kidney disease (CKD) models by sodium-glucose cotransporter 2 (SGLT2) inhibitors are not well defined. Five groups were established: sham-operated rats, placebo-treated rats with 5/6 nephrectomy (5/6Nx), 5/6Nx + telmisartan (5 mg/kg/day), 5/6Nx + empagliflozin (3 mg/kg/day), and 5/6Nx + empagliflozin (15 mg/kg/day). Treatment duration was 95 days. Empagliflozin showed a dose-dependent beneficial effect on the change from baseline of creatinine clearance (Ccr). The urinary albumin-to-creatinine ratio likewise improved in a dose-dependent manner. Both dosages of empagliflozin improved morphological kidney damage parameters such as renal interstitial fibrosis and glomerulosclerosis. 5/6 nephrectomy led to a substantial reduction of urinary adenosine excretion, a surrogate parameter of the tubuloglomerular feedback (TGF) mechanism. Empagliflozin caused a dose-dependent increase in urinary adenosine excretion. The urinary adenosine excretion was negatively correlated with renal interstitial fibrosis and positively correlated with Ccr. Immunofluorescence analysis revealed that empagliflozin had no effect on CD8+ and CD4+ T cells as well as on CD68+ cells (macrophages). To further explore potential mechanisms, a nonhypothesis-driven approach was used. RNA sequencing followed by quantitative real-time polymerase chain reaction revealed that complement component 1Q subcomponent A chain (C1QA) as well as complement component 1Q subcomponent C chain (C1QC) gene expression were upregulated in the placebo-treated 5/6Nx rats and this upregulation was blunted by treatment with empagliflozin. In conclusion, empagliflozin-mediated nephroprotection in nondiabetic CKD is due to a dose-dependent activation of the TGF as well as empagliflozin-mediated effects on the complement system.


Subject(s)
Diabetes Mellitus, Type 2 , Renal Insufficiency, Chronic , Sodium-Glucose Transporter 2 Inhibitors , Rats , Animals , Complement C1q , Creatinine , Feedback , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Fibrosis
3.
Kidney Blood Press Res ; 47(9): 565-575, 2022.
Article in English | MEDLINE | ID: mdl-35878596

ABSTRACT

INTRODUCTION: The angiotensin-converting enzyme 2 (ACE2) as well as the transmembrane protease serine type 2 (TMPRSS2) have been found to play roles in cell entry for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus causing coronavirus disease 2019 (COVID-19). SARS-CoV-2 infection risk and severity of COVID-19 might be indicated by the expression of ACE2 and TMPRSS2 in the lung. METHODS: A high-salt diet rat model and renin-angiotensin-aldosterone system (RAAS) blockade were used to test whether these factors affect ACE2 and TMPRSS2 expression in the lung. A normal (0.3% NaCl), a medium (2% NaCl), or a high (8% NaCl) salt diet was fed to rats for 12 weeks, along with enalapril or telmisartan, before examining the lung for histopathological alteration. Using immunofluorescence and qRT-PCR, the localization as well as mRNA expression of ACE2 and TMPRSS2 were investigated. RESULTS: The findings provide evidence that both TMPRSS2 and ACE2 are highly expressed in bronchial epithelial cells as well as ACE2 was also expressed in alveolar type 2 cells. High-salt diet exposure in rats leads to elevated ACE2 expression on protein level. Treatment with RAAS blockers had no effect on lung tissue expression of ACE2 and TMPRSS2. CONCLUSIONS: These findings offer biological support regarding the safety of these drugs that are often prescribed to COVID-19 patients with cardiovascular comorbidity. High salt intake, on the other hand, might adversely affect COVID-19 outcome. Our preclinical data should stimulate clinical studies addressing this point of concern.


Subject(s)
COVID-19 , Renin-Angiotensin System , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , Animals , Enalapril/pharmacology , Lung , RNA, Messenger/metabolism , Rats , Renin-Angiotensin System/drug effects , Serine Endopeptidases , Sodium Chloride, Dietary/adverse effects , Telmisartan/pharmacology
4.
BMC Nephrol ; 23(1): 117, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35331159

ABSTRACT

BACKGROUND: Host factors such as angiotensin-converting enzyme 2 (ACE2) and the transmembrane protease, serine-subtype-2 (TMPRSS2) are important factors for SARS-CoV-2 infection. Clinical and pre-clinical studies demonstrated that RAAS-blocking agents can be safely used during a SARS-CoV-2 infection but it is unknown if DPP-4 inhibitors or SGLT2-blockers may promote COVID-19 by increasing the host viral entry enzymes ACE2 and TMPRSS2. METHODS: We investigated telmisartan, linagliptin and empagliflozin induced effects on renal and cardiac expression of ACE2, TMPRSS2 and key enzymes involved in RAAS (REN, AGTR2, AGT) under high-salt conditions in a non-diabetic experimental 5/6 nephrectomy (5/6 Nx) model. In the present study, the gene expression of Ace2, Tmprss2, Ren, Agtr2 and Agt was assessed with qRT-PCR and the protein expression of ACE2 and TMPRSS2 with immunohistochemistry in the following experimental groups: Sham + normal diet (ND) + placebo (PBO); 5/6Nx + ND + PBO; 5/6Nx + high salt-diet (HSD) + PBO; 5/6Nx + HSD + telmisartan; 5/6Nx + HSD + linagliptin; 5/6Nx + HSD + empagliflozin. RESULTS: In the kidney, the expression of Ace2 was not altered on mRNA level under disease and treatment conditions. The renal TMPRSS2 levels (mRNA and protein) were not affected, whereas the cardiac level was significantly increased in 5/6Nx rats. Intriguingly, the elevated TMPRSS2 protein expression in the heart was significantly normalized after treatment with telmisartan, linagliptin and empagliflozin. CONCLUSIONS: Our study indicated that there is no upregulation regarding host factors potentially promoting SARS-CoV-2 virus entry into host cells when the SGLT2-blocker empagliflozin, telmisartan and the DPP4-inhibitor blocker linagliptin are used. The results obtained in a preclinical, experimental non-diabetic kidney failure model need confirmation in ongoing interventional clinical trials.


Subject(s)
COVID-19 Drug Treatment , Dipeptidyl-Peptidase IV Inhibitors , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors , Animals , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Humans , Kidney/metabolism , Nephrectomy , Rats , SARS-CoV-2 , Sodium-Glucose Transporter 2 , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism
5.
Respir Res ; 22(1): 158, 2021 May 22.
Article in English | MEDLINE | ID: mdl-34022896

ABSTRACT

BACKGROUND: RORγt is a transcription factor that enables elaboration of Th17-associated cytokines (including IL-17 and IL-22) and is proposed as a pharmacological target for severe asthma. METHODS: IL-17 immunohistochemistry was performed in severe asthma bronchial biopsies (specificity confirmed with in situ hybridization). Primary human small airway epithelial cells in air liquid interface and primary bronchial smooth muscle cells were stimulated with recombinant human IL-17 and/or IL-22 and pro-inflammatory cytokines measured. Balb/c mice were challenged intratracheally with IL-17 and/or IL-22 and airway hyperreactivity, pro-inflammatory cytokines and airway neutrophilia measured. Balb/c mice were sensitized intraperitoneally and challenged intratracheally with house dust mite extract and the effect of either a RORγt inhibitor (BIX119) or an anti-IL-11 antibody assessed on airway hyperreactivity, pro-inflammatory cytokines and airway neutrophilia measured. RESULTS: We confirmed in severe asthma bronchial biopsies both the presence of IL-17-positive lymphocytes and that an IL-17 transcriptome profile in a severe asthma patient sub-population. Both IL-17 and IL-22 stimulated the release of pro-inflammatory cytokine and chemokine release from primary human lung cells and in mice. Furthermore, IL-22 in combination with IL-17, but neither alone, elicits airway hyperresponsiveness (AHR) in naïve mice. A RORγt inhibitor specifically blocked both IL-17 and IL-22, AHR and neutrophilia in a mouse house dust mite model unlike other registered or advanced pipeline modes of action. Full efficacy versus these parameters was associated with 90% inhibition of IL-17 and 50% inhibition of IL-22. In contrast, anti-IL-17 also blocked IL-17, but not IL-22, AHR or neutrophilia. Moreover, the deregulated genes in the lungs from these mice correlated well with deregulated genes from severe asthma biopsies suggesting that this model recapitulates significant severe asthma-relevant biology. Furthermore, these genes were reversed upon RORγt inhibition in the HDM model. Cell deconvolution suggested that the responsible cells were corticosteroid insensitive γδ-T-cells. CONCLUSION: These data strongly suggest that both IL-17 and IL-22 are required for Th2-low endotype associated biology and that a RORγt inhibitor may provide improved clinical benefit in a severe asthma sub-population of patients by blocking both IL-17 and IL-22 biology compared with blocking IL-17 alone.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Interleukin-17/metabolism , Interleukins/antagonists & inhibitors , Lung/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Th17 Cells/drug effects , Adolescent , Adult , Aged , Animals , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Humans , Interleukins/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Male , Mice, Inbred BALB C , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pyroglyphidae/immunology , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult , Interleukin-22
6.
Diabetes Obes Metab ; 23(8): 1968-1972, 2021 08.
Article in English | MEDLINE | ID: mdl-33881796

ABSTRACT

Results of a post hoc analysis of urinary dipeptidyl peptidase-4 (DPP-4) protein as a predictor of urine albumin-to-creatinine ratio (UACR) response to linagliptin treatment based on MARLINA-T2D trial data are described. MARLINA was a 24-week, phase 3b, multinational, placebo-controlled clinical trial, in which patients with type 2 diabetes (T2D), HbA1c 6.5%-10.0% and UACR 30-3000 mg/g (n = 360) were treated with linagliptin or placebo. After 24 weeks of treatment, linagliptin significantly inhibited urinary DPP-4 activity and increased urinary DPP-4 protein. Furthermore, medium urinary DPP-4 protein levels (between 5.5 and 7.5 natural logarithmic [ln] µg/g creatinine) at baseline allowed for prediction of improved UACR in linagliptin-treated individuals. In patients with lower or higher levels of urinary DPP-4 protein at baseline, no association between linagliptin treatment and improved UACR was present. This might suggest a varying degree of importance of DPP-4 as a pathophysiological factor in T2D-associated kidney disease. In summary, urinary DPP-4 might be a useful predictive biomarker for UACR improvement by linagliptin.


Subject(s)
Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Albumins , Biomarkers , Creatinine , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4 , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Humans , Linagliptin/therapeutic use
7.
Malar J ; 19(1): 49, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31996238

ABSTRACT

BACKGROUND: Vaccination induces survival of otherwise lethal blood-stage infections of the experimental malaria Plasmodium chabaudi. Blood-stage malaria induces extramedullary erythropoiesis in the liver. This study investigates how vaccination affects the course of malaria-induced expression of erythrocytic genes in the liver. METHODS: Female Balb/c mice were vaccinated at week 3 and week 1 before challenging with 106 P. chabaudi-parasitized erythrocytes. The non-infectious vaccine consisted of erythrocyte ghosts isolated from P. chabaudi-infected erythrocytes. Gene expression microarrays and quantitative real-time PCR were used to compare mRNA expression of different erythrocytic genes in the liver of vaccination-protected and non-protected mice during infections on days 0, 1, 4, 8, and 11 p.i. RESULTS: Global transcriptomics analyses reveal vaccination-induced modifications of malaria-induced increases in hepatic gene expression on days 4 and 11 p.i. On these days, vaccination also alters hepatic expression of the erythropoiesis-involved genes Ermap, Kel, Rhd, Rhag, Slc4a1, Gypa, Add2, Ank1, Epb4.1, Epb4.2, Epb4.9, Spta1, Sptb, Tmod1, Ahsp, Acyp1, Gata1, Gfi1b, Tal1, Klf1, Epor, and Cldn13. In vaccination-protected mice, expression of these genes, except Epb4.1, is significantly higher on day 4 p.i. than in un-protected non-vaccinated mice, reaches maximal expression at peak parasitaemia on day 8 p.i., and is slowed down or even decreased towards the end of crisis phase on day 11 p.i.. After day 1 p.i., Epor expression takes about the same course as that of the other erythroid genes. Hepatic expression of Epo, however, is delayed in both vaccinated and non-vaccinated mice for the first 4 days p.i. and is maximal at significantly higher levels in vaccinated mice on day 8 p.i., before declining towards the end of crisis phase on day 11 p.i. CONCLUSION: The present data indicate that vaccination accelerates malaria-induced erythroblastosis in the liver for 1-2 days. This may contribute to earlier replenishment of peripheral red blood cells by liver-derived reticulocytes, which may favour final survival of otherwise lethal blood-stage malaria, since reticulocytes are not preferred as host cells by P. chabaudi.


Subject(s)
Erythropoiesis/immunology , Liver/pathology , Malaria/blood , Plasmodium chabaudi/immunology , Vaccination/adverse effects , Animals , Erythrocyte Membrane/immunology , Erythropoiesis/genetics , Female , Liver/parasitology , Malaria/pathology , Malaria Vaccines/adverse effects , Mice , Mice, Inbred BALB C , Principal Component Analysis , Real-Time Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Transcriptome
8.
Int J Mol Sci ; 22(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396535

ABSTRACT

MicroRNAs (miRNAs) are short, non-coding RNA species that are important post-transcriptional regulators of gene expression and play an important role in the pathogenesis of non-alcoholic fatty liver disease. Here, we investigated the phosphodiesterase 5 (PDE5) inhibitor induced effects on hepatic and plasma exosomal miRNA expression in CCl4-treated rats. In the present study, hepatic miRNA profiling was conducted using the Nanostring nCounter technology and mRNA profiling using RNA sequencing from PDE5 treated rats in the model of CCl4-induced liver fibrosis. To evaluate if the PDE5 inhibitor affected differentially expressed miRNAs in the liver can be detected in plasma exosomes, qRT-PCR specific assays were used. In livers from CCl4-treated rats, the expression of 22 miRNAs was significantly increased (> 1.5-fold, adj. p < 0.05), whereas the expression of 16 miRNAs was significantly decreased (> 1.5-fold, adj. p < 0.05). The majority of the deregulated miRNA species are implicated in fibrotic and inflammatory processes. The PDE5 inhibitor suppressed the induction of pro-fibrotic miRNAs, such as miR-99b miR-100 and miR-199a-5p, and restored levels of anti-fibrotic miR-122 and miR-192 in the liver. In plasma exosomes, we observed elevated levels of miR-99b, miR-100 and miR-142-3p after treatment with the PDE5-inhibitor compared to CCl4/Vehicle-treated. Our study demonstrated for the first time that during the development of hepatic fibrosis in the preclinical model of CCl4-induced liver fibrosis, defined aspects of miRNA regulated liver pathogenesis are influenced by PDE5 treatment. In conclusion, miRNA profiling of plasma exosomes might be used as a biomarker for NASH progression and monitoring of treatment effects.


Subject(s)
Biomarkers/analysis , Carbon Tetrachloride/toxicity , Exosomes/genetics , Gene Expression Regulation/drug effects , Liver Cirrhosis/drug therapy , MicroRNAs/genetics , Phosphodiesterase 5 Inhibitors/pharmacology , Animals , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA
9.
Kidney Int ; 95(6): 1373-1388, 2019 06.
Article in English | MEDLINE | ID: mdl-30979564

ABSTRACT

Dipeptidyl peptidase type 4 (DPP-4) inhibitors were reported to have beneficial effects in experimental models of chronic kidney disease. The underlying mechanisms are not completely understood. However, these effects could be mediated via the glucagon-like peptide-1 (GLP-1)/GLP-1 receptor (GLP1R) pathway. Here we investigated the renal effects of the DPP-4 inhibitor linagliptin in Glp1r-/- knock out and wild-type mice with 5/6 nephrectomy (5/6Nx). Mice were allocated to groups: sham+wild type+placebo; 5/6Nx+ wild type+placebo; 5/6Nx+wild type+linagliptin; sham+knock out+placebo; 5/6Nx+knock out+ placebo; 5/6Nx+knock out+linagliptin. 5/6Nx caused the development of renal interstitial fibrosis, significantly increased plasma cystatin C and creatinine levels and suppressed renal gelatinase/collagenase, matrix metalloproteinase-1 and -13 activities; effects counteracted by linagliptin treatment in wildtype and Glp1r-/- mice. Two hundred ninety-eight proteomics signals were differentially regulated in kidneys among the groups, with 150 signals specific to linagliptin treatment as shown by mass spectrometry. Treatment significantly upregulated three peptides derived from collagen alpha-1(I), thymosin ß4 and heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) and significantly downregulated one peptide derived from Y box binding protein-1 (YB-1). The proteomics results were further confirmed using western blot and immunofluorescence microscopy. Also, 5/6Nx led to significant up-regulation of renal transforming growth factor-ß1 and pSMAD3 expression in wild type mice and linagliptin significantly counteracted this up-regulation in wild type and Glp1r-/- mice. Thus, the renoprotective effects of linagliptin cannot solely be attributed to the GLP-1/GLP1R pathway, highlighting the importance of other signaling pathways (collagen I homeostasis, HNRNPA1, YB-1, thymosin ß4 and TGF-ß1) influenced by DPP-4 inhibition.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Kidney/drug effects , Linagliptin/pharmacology , Renal Insufficiency, Chronic/drug therapy , Signal Transduction/drug effects , Animals , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Disease Models, Animal , Down-Regulation/drug effects , Glucagon-Like Peptide-1 Receptor/genetics , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Humans , Kidney/pathology , Kidney/surgery , Linagliptin/therapeutic use , Male , Mice , Mice, Knockout , Nephrectomy/adverse effects , RNA-Seq , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology , Thymosin/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation/drug effects
10.
Microb Pathog ; 117: 299-303, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29496525

ABSTRACT

Malaria is a harmful disease affecting both tropical and subtropical countries and causing sometimes fatal complications. The effects of malaria-related complications on the intestine have been relatively neglected, and the reasons for the intestinal damage caused by malaria infection are not yet clear. The present study aims to evaluate the influence of intestinal vitamin D receptor on host-pathogen interactions during malaria induced in mice by Plasmodium chabaudi. To induce the infection, animals were infected with 106P. chabaudi-parasitized erythrocytes. Mice were sacrificed on day 8 post-infection. The infected mice experienced a significant body weight loss and parasitaemia affecting about 46% of RBCs. Infection caused marked pathological changes in the intestinal tissue indicated by shortening of the intestine and villi. Moreover, the phagocytic activity of macrophages increased significantly (P < 0.01) in the infected villi compared to the non-infected ones. Infection by the parasite also induced marked upregulation of nuclear factor-kappa B, inducible nitric oxide synthase, Vitamin D Receptor, interleukin-1ß, tumour necrosis factor alpha and interferon gamma-mRNA. It can be implied from this that vitamin D receptor has a role in regulating malarial infection.


Subject(s)
Host-Parasite Interactions/physiology , Intestinal Mucosa/metabolism , Malaria/blood , Malaria/complications , Plasmodium chabaudi/pathogenicity , Receptors, Calcitriol/physiology , Animals , Body Weight , Disease Models, Animal , Erythrocytes/parasitology , Erythrocytes/pathology , Female , Gene Expression Regulation , Host-Parasite Interactions/genetics , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Intestines/parasitology , Intestines/pathology , Macrophages/metabolism , Malaria/parasitology , Malaria/pathology , Mice , Mice, Inbred C57BL , NF-kappa B p50 Subunit/metabolism , Nitric Oxide Synthase Type II/metabolism , Parasitemia , Phagocytosis , RNA, Messenger/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
11.
Malar J ; 17(1): 215, 2018 May 29.
Article in English | MEDLINE | ID: mdl-29843710

ABSTRACT

BACKGROUND: The role of the liver for survival of blood-stage malaria is only poorly understood. In experimental blood-stage malaria with Plasmodium chabaudi, protective vaccination induces healing and, thus, survival of otherwise lethal infections. This model is appropriate to study the role of the liver in vaccination-induced survival of blood-stage malaria. METHODS: Female Balb/c mice were vaccinated with a non-infectious vaccine consisting of plasma membranes isolated in the form of erythrocyte ghosts from P. chabaudi-infected erythrocytes at week 3 and week 1 before infection with P. chabaudi blood-stage malaria. Gene expression microarrays and quantitative real-time PCR were used to investigate the response of the liver, in terms of expression of mRNA and long intergenic non-coding (linc)RNA, to vaccination-induced healing infections and lethal P. chabaudi malaria at early patency on day 4 post infection, when parasitized erythrocytes begin to appear in peripheral blood. RESULTS: In vaccination-induced healing infections, 23 genes were identified to be induced in the liver by > tenfold at p < 0.01. More than one-third were genes known to be involved in erythropoiesis, such as Kel, Rhag, Ahsp, Ermap, Slc4a1, Cldn13 Gata1, and Gfi1b. Another group of > tenfold expressed genes include genes involved in natural cytotoxicity, such as those encoding killer cell lectin-like receptors Klrb1a, Klrc3, Klrd1, the natural cytotoxicity-triggering receptor 1 Ncr1, as well as the granzyme B encoding Gzmb. Additionally, a series of genes involved in the control of cell cycle and mitosis were identified: Ccnb1, Cdc25c, Ckap2l were expressed > tenfold only in vaccination-protected mice, and the expression of 22 genes was at least 100% higher in vaccination-protected mice than in non-vaccinated mice. Furthermore, distinct lincRNA species were changed by > threefold in livers of vaccination-protected mice, whereas lethal malaria induced different lincRNAs. CONCLUSION: The present data suggest that protective vaccination accelerates the malaria-induced occurrence of extramedullary erythropoiesis, generation of liver-resident cytotoxic cells, and regeneration from malaria-induced injury in the liver at early patency, which may be critical for final survival of otherwise lethal blood-stage malaria of P. chabaudi.


Subject(s)
Gene Expression , Malaria Vaccines/immunology , Malaria/genetics , Plasmodium chabaudi/physiology , Animals , Female , Liver/metabolism , Liver/parasitology , Malaria/immunology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Specific Pathogen-Free Organisms
12.
Parasitol Res ; 117(4): 1115-1129, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29404749

ABSTRACT

Current knowledge about liver responses to blood-stage malaria and their modulation by vaccination is still unclear. This study investigated effects of protective vaccination on liver gene and lincRNA expression of Balb/c mice at early prepatency of Plasmodium chabaudi blood-stage malaria. When a blood-stage vaccine was used to induce > 80% survival of otherwise lethal malaria, significant differences (p < 0.01) were detectable in global liver gene expression between vaccination-protected (potentially surviving) and non-protected non-vaccinated mice on day 1 p.i.. In the livers of protected mice, gene expression microarrays identified 224 and 419 genes, whose expression was up- and downregulated by > 3-fold, respectively. There were 24 genes upregulated by > 10-fold, including 10 IFN-inducible genes encompassing GTPases Irgm1, 2, and 3, and guanylate-binding protein Gbp11, the IL-1 decoy receptors Il1f9 and Il1ra1, the Il6 gene, and the gene for facilitated glucose transportation. Moreover, the IL-18 decoy receptor gene Il18bp, Gzmb, the genes Lif and Osmr encoding proteins of the IL-6 family, and the taurine transporter gene Slc6a6 were expressed > 3-fold in vaccinated mice. The genes Gbp10, 6, 4 were expressed by > 50% in vaccination-protected than in non-vaccinated mice. In addition, 43 lincRNA species were up- and 36 downregulated. Our data suggested novel regulatory elements of potential anti-malaria activity activated by protective vaccination in the liver, evidenced in response to early prepatent infections in vaccination-protected mice of otherwise lethal blood-stage malaria of P. chabaudi.


Subject(s)
Gene Expression Regulation/genetics , Liver/metabolism , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium chabaudi/immunology , RNA, Long Noncoding/biosynthesis , Animals , Down-Regulation/genetics , Female , Gene Expression , Liver/parasitology , Malaria/parasitology , Mice , Mice, Inbred BALB C , Microarray Analysis , RNA, Long Noncoding/genetics , Up-Regulation/genetics , Vaccination
13.
Parasitol Res ; 115(5): 1835-43, 2016 May.
Article in English | MEDLINE | ID: mdl-26809341

ABSTRACT

Protective vaccination induces self-healing of otherwise lethal blood-stage infections of Plasmodium chabaudi malaria. Here, we investigate mRNA expression patterns of all 12 members of the Toll-like receptor (Tlr) gene family in the liver, a major effector organ against blood-stage malaria, during lethal and vaccination-induced self-healing infections of P. chabaudi in female Balb/c mice. Gene expression microarrays reveal that all 12 Tlr genes are constitutively expressed, though at varying levels, and specifically respond to infection. Protective vaccination does not affect constitutive expression of any of the 12 Tlr genes but leads to differential expression (p < 0.05) of seven Tlrs (1, 2, 4, 7, 8, 12, and 13) in response to malaria. Quantitative PCR substantiates differential expression at p < 0.01. There is an increased expression of Tlr2 by approximately five-fold on day 1 post-infection (p.i.) and Tlr1 by approximately threefold on day 4 p.i.. At peak parasitemia on day 8 p.i., none of the 12 Tlrs display any differential expression. After peak parasitemia, towards the end of the crisis phase on day 11 p.i., expression of Tlrs 1, 4, and 12 is increased by approximately four-, two-, and three-fold, respectively, and that of Tlr7 is decreased by approximately two-fold. Collectively, our data suggest that though all 12 members of the Tlr gene family are specifically responsive to malaria in the liver, not only Tlr2 at the early stage of infection but also the Tlrs 1, 4, 7, and 12 towards the end of crisis phase are critical for vaccination-induced resolution and survival of otherwise lethal blood-stage malaria.


Subject(s)
Liver/metabolism , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium chabaudi , Toll-Like Receptors/metabolism , Animals , Female , Gene Expression Regulation , Liver/immunology , Malaria/immunology , Malaria/metabolism , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microarray Analysis , Parasitemia , Toll-Like Receptors/genetics , Vaccination
14.
Int J Mol Sci ; 17(5)2016 May 03.
Article in English | MEDLINE | ID: mdl-27153060

ABSTRACT

The aim of the present pilot study was the identification of micro-RNA changes over time during the development and progression of type 2 diabetes (T2D) in Zucker diabetic fatty rats (ZDF rats). T2D is a complex metabolic disorder that is characterized, inter alia, by progressive failure of pancreatic ß cells to produce insulin, but also by functional or morphological modifications of others organ, such as liver, adipose tissue and the cardiovascular system. Micro-RNAs are a novel class of biomarkers that have the potential to represent biomarkers of disease progression. In this study, the onset and progression of diabetes was followed in ZDF rats from six weeks until 17 weeks of age. After an initial phase of hyperinsulinemia, the animals developed T2D and lost the capacity to produce sufficient insulin. Circulating miRNAs were measured from plasma samples at four time points: pre-diabetes (six weeks of age), hyperinsulinemia (eight weeks), ß cell failure (11 weeks) and late-stage diabetes (17 weeks) using TaqMan miRNA arrays. Bioinformatic analysis revealed distinct changes of circulating miRNAs over time. Several miRNAs were found to be increased over the course of the disease progression, such as miR-122, miR-133, miR-210 and miR-375. The most significantly decreased miRNAs were miR-140, miR-151-3p, miR-185, miR-203, miR-434-3p and miR-450a. Some of the miRNAs have also been identified in type 2 diabetic patients recently and, therefore, may have the potential to be useful biomarkers for the disease progression of T2D and/or the treatment response for anti-diabetic medications.


Subject(s)
Diabetes Mellitus, Type 2/blood , MicroRNAs/blood , Animals , Biomarkers/blood , Insulin/blood , Male , Rats , Rats, Zucker
15.
Parasitol Res ; 114(10): 3711-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26122996

ABSTRACT

Schistosomiasis is a condition characterized by high rates of morbidity and cognitive impairment. It afflicts many people in tropical and sub-tropical countries. Our study aimed to investigate the protective role of gold nanoparticles (GNPs) on the brain of mice infected with Schistosoma mansoni. Characterizations of GNPs were determined by using high-resolution transmission electron microscopy. Three doses of GNPs (0.25, 0.5, and 1.0 mg/kg body weight) were used to treat animals after S. mansoni infection. The infection induced impairments in histological picture as a result of schistosome infection resulting in a disturbance in the content of the brain neurotransmitters, norepinephrine (NE), and dopamine (DA). Also, the infection induced significant reduction in glutathione level; oppositely, the levels of nitric oxide and malondialdehyde were increased significantly. In addition, S. mansoni was able to disregulate the infected mice brain Cacnb4, Cabp4, Vdac3, Glrb, and Adam23 messenger RNA (mRNA). On the other hand, treatment of mice with GNPs could alleviate the histological impairments, the changes in the content of NE and DA, and the brain oxidative damage. Also, GNPs could regulate the gene expression due to S. mansoni infection. Generally, GNPs could decrease the neurooxidative stress and regulated the gene expression in the brain of infected mice. Consequently, our results revealed an anti-neuroschistosomal effect of GNPs in mice infected with S. mansoni.


Subject(s)
Brain/parasitology , Gold/therapeutic use , Metal Nanoparticles/therapeutic use , Schistosoma mansoni/drug effects , Schistosomiasis mansoni/drug therapy , Animals , Brain/pathology , Glutathione/metabolism , Gold/chemistry , Male , Malondialdehyde/metabolism , Metal Nanoparticles/chemistry , Mice
16.
Parasitol Res ; 114(4): 1581-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25663104

ABSTRACT

Plant-based natural products are promising sources for identifying novel agents with potential anti-Eimeria activity. This study explores possible effects of berberine on Eimeria papillata infections in the jejunum of male Swiss albino mice. Berberine chloride, when daily administered to mice during infection, impairs intracellular development and multiplication of E. papillata, evidenced as 60% reduction of maximal fecal output of oocysts on day 5 p.i. Concomitantly, berberine attenuates the inflammatory response, evidenced as decreased messenger RNA (mRNA) expression of IL-1ß, IL-6, TNFα, IFNγ, and iNOS, as well as the oxidative stress response, evidenced as impaired increase in malondialdehyde, nitrate, and H2O2 and as prevented decrease in glutathione and catalase activity. Berberine also alters gene expression in the infected jejunum. On day 5 p.i., mRNA expression of 29 genes with annotated functions is more than 10-fold upregulated and that of 14 genes downregulated. Berberine downregulates the genes Xaf1, Itgb3bp, and Faim3 involved in apoptotic processes and upregulates genes involved in innate immune responses, as e.g., Colec11, Saa2, Klra8, Clec1b, and Crtam, especially the genes Cpa3, Fcer1a, and Mcpt1, Mcpt2, and Mcpt4 involved in mast cell activity. Additionally, 18 noncoding lincRNA species are differentially expressed more than 10-fold under berberine. Our data suggest that berberine induces hosts to exert anti-Eimeria activity by attenuating the inflammatory and oxidative stress response, by impairing apoptotic processes, and by activating local innate immune responses and epigenetic mechanisms in the host jejunum. Berberine has the potential as an anti-Eimeria food additive in animal farming.


Subject(s)
Antiprotozoal Agents/pharmacology , Berberine/pharmacology , Coccidiosis/drug therapy , Coccidiosis/genetics , Eimeria/drug effects , Jejunum/parasitology , Animals , Apoptosis/drug effects , Coccidiosis/metabolism , Coccidiosis/parasitology , Eimeria/physiology , Gene Expression/drug effects , Glutathione/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Jejunum/metabolism , Male , Malondialdehyde/metabolism , Mice , Oxidative Stress/drug effects
17.
J Immunol ; 188(9): 4141-4, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22467660

ABSTRACT

Circulating IL-6 levels correlate with the severity of blood-stage malaria in humans and mouse models, but the impact of IL-6 classic signaling through membrane IL-6Rα, as well as IL-6 trans-signaling through soluble IL-6Rα, on the outcome of malaria has remained unknown. In this study, we created IL-6Rα-deficient mice that exhibit a 50% survival of otherwise lethal blood-stage malaria of the genus Plasmodium chabaudi. Inducing IL-6 trans-signaling by injection of mouse recombinant soluble IL-6Rα in IL-6Rα-deficient mice restores the lethal outcome to malaria infection. In contrast, inhibition of IL-6 trans-signaling via injection of recombinant sGP130Fc protein in control mice results in a 40% survival rate. Our data demonstrate that IL-6 trans-signaling, rather than classic IL-6 signaling, contributes to malaria-induced lethality in mice, preceded by an increased inflammatory response. Therefore, inhibition of IL-6 trans-signaling may serve as a novel promising therapeutic basis to combat malaria.


Subject(s)
Interleukin-6/immunology , Malaria/immunology , Plasmodium chabaudi/immunology , Signal Transduction/immunology , Animals , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/immunology , Cytokine Receptor gp130/pharmacology , Interleukin-6/genetics , Interleukin-6 Receptor alpha Subunit/genetics , Interleukin-6 Receptor alpha Subunit/immunology , Malaria/genetics , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Parasitol Res ; 113(10): 3609-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25056943

ABSTRACT

Testosterone (T) is known to induce persistent susceptibility to Plasmodium chabaudi malaria. Pathogens recognizing Toll-like receptors (TLRs), though potentially important against malaria, have not yet been examined for their T-sensitivity. Here, we investigate effects of T and P. chabaudi on mRNA expression and promoter DNA methylation of Tlr1-9 genes in the liver of female C57BL/6 mice. These are treated with T or vehicle for 3 weeks, and then treatment is discontinued for 12 weeks, before challenging with P. chabaudi for 8 days. Our data reveal that T induces a 9.1-fold downregulation of Tlr6 mRNA and 6.3-fold upregulation of Tlr8 mRNA. Blood-stage infections induce significant increases in mRNA expression of Tlr1, 2, 4, 6, 7, and 8 varying between 2.5-fold and 21-fold in control mice. In T-pretreated mice, these Tlr genes are also significantly responsive to infections. However, the malaria-induced upregulations of the relative mRNA expressions of Tlr6 and Tlr8 are 5.6-fold higher and 6.5-fold lower in T-pretreated mice than in control mice. Infections induce a massive DNA down-methylation of the Tlr6 gene promoter in control mice, which is still more pronounced in T-pretreated mice, while significant changes are not detectable for the DNA methylation status of the Tlr8 promoter. Our data support the view that hepatic expression of Tlr6, but not that of Tlr8 is epigenetically controlled, and that the dysregulations of Tlr6 and Tlr8 critically contribute to T-induced persistent susceptibility to P. chabaudi malaria, possibly by dys-balancing responses of TLR6-mediated pathogen recognition and TLR8-mediated generation of anti-malaria "protective" autoimmunity.


Subject(s)
Malaria/metabolism , Plasmodium chabaudi/immunology , Promoter Regions, Genetic/genetics , Testosterone/pharmacology , Toll-Like Receptor 6/genetics , Toll-Like Receptor 8/genetics , Animals , DNA Methylation/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation , Liver/drug effects , Liver/metabolism , Malaria/immunology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Specific Pathogen-Free Organisms , Up-Regulation
19.
Microsc Res Tech ; 87(10): 2437-2446, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38845567

ABSTRACT

Coccidiosis poses significant hazards to animals, particularly in terms of compromised health, reduced productivity, and economic losses in livestock farming. The conventional treatments for coccidiosis often involve synthetic drugs, contributing to concerns about drug resistance and environmental impact. The pressing need for eco-friendly alternatives is highlighted in this study, emphasizing the importance of exploring medicinal plants like Cassia alata leaf extracts (CAE) against Eimeria papillata-induced infection in mice. The CAE exhibited significant phenolic (2.17 ± 0.03 g/100 g) and flavonoid (0.14 ± 0.01 g/100 g) content and demonstrated notable antioxidant activity. In infected mice, the CAE treatment led to a substantial reduction in oocyst output (~6 fold), ameliorating necrotic enteritis and inflammatory changes in the jejunum. Additionally, CAE treatment increased goblet cell numbers (9.3 ± 0.1 / villus) and decreased macrophage infiltration in the intestinal villi. Molecular analyses revealed CAE's positive modulation of MUC2 gene and notably reduced the levels of pro-inflammatory cytokines (specifically IL-1ß, IL-10, and IFN-γ) when contrasted with the infected cohort. Furthermore, CAE treatment significantly reduced nitric oxide levels (44.03 ± 2.4 µmol/mg), showcasing its anti-inflammatory properties. The findings of this study not only contribute to the understanding of CAE's therapeutic potential but also underscore the importance of seeking eco-friendly alternatives in the face of coccidiosis challenges, addressing both the well-being of animals and the sustainability of agricultural practices. RESEARCH HIGHLIGHTS: Cassia alata extract (CAE) exhibited significant phenolic and flavonoid content, displaying notable antioxidant activity. In infected mice, CAE treatment led to a substantial reduction in oocyst output, ameliorating necrotic enteritis and inflammatory changes in the jejunum. CAE treatment increased goblet cell numbers and decreased macrophage infiltration in the intestinal villi, while molecular analyses revealed its positive modulation of the MUC2 gene and notable reduction in pro-inflammatory cytokine levels. Additionally, CAE treatment significantly reduced nitric oxide levels, showcasing its anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents , Cassia , Coccidiosis , Cytokines , Eimeria , Jejunum , Mucin-2 , Plant Extracts , Animals , Jejunum/parasitology , Jejunum/drug effects , Jejunum/pathology , Coccidiosis/drug therapy , Coccidiosis/veterinary , Coccidiosis/parasitology , Mice , Plant Extracts/pharmacology , Eimeria/drug effects , Cassia/chemistry , Anti-Inflammatory Agents/pharmacology , Mucin-2/metabolism , Mucin-2/genetics , Cytokines/metabolism , Plant Leaves/chemistry , Disease Models, Animal
20.
Article in English | MEDLINE | ID: mdl-39360533

ABSTRACT

INTRODUCTION: The concurrent presence of helminthiasis and bacterial diseases imposes a dual burden, worsening the challenges associated with each condition independently. This cohabitation intensifies the economic impact, creating a compounding effect on public health and economic well-being. METHOD: Phytochemical analysis of Cassia alata Extract (CAE) using infrared spectroscopy has revealed the presence of various functional groups. In addition, GC mass analysis has confirmed the presence of 26 active compounds. An assessment of the anthelmintic activity of CAE against mature earthworms has demonstrated comparable efficacy to the conventional anthelmintic, albendazole. The optimal dosage of 500 mg/ml has induced a rapid onset of paralysis (2.7 ± 0.5 min) and death (20.1 ± 1.7 min), outperforming albendazole (20 mg/mL) in terms of faster paralysis and death times (21.8 ± 1.1 and 30.14 ± 3.2 min, respectively). Structural modifications induced by CAE have been observed through light microscopy and Scanning Electron Microscopy (SEM). Control worms have exhibited normal body architecture, while CAE-treated worms have displayed size reduction, uniform body wall shrinkage, and increased cuticular thickness. Similar alterations have been observed in albendazole-treated worms. RESULTS: The antibacterial activity of CAE has been evaluated through a broth dilution assay, which has revealed a dose-response effect. At 6.25 mg/ml, CAE has exhibited 100% inhibitory action against both Gram-positive and Gram-negative bacteria. Significant differences in bacterial viability have been noted at lower concentrations, with no significant variation at 0.3906 mg/ml of CAE. CONCLUSION: The findings have highlighted the multifaceted bioactivity of CAE, showcasing its potential as an anthelmintic agent and antimicrobial agent against a spectrum of bacterial strains. The observed structural alterations in treated worms have provided insights into the potential mechanisms underlying the anthelmintic effects.

SELECTION OF CITATIONS
SEARCH DETAIL