Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Front Immunol ; 14: 1252818, 2023.
Article in English | MEDLINE | ID: mdl-37809105

ABSTRACT

Introduction: Hemolytic-uremic syndrome (HUS) can occur as a systemic complication of infection with Shiga toxin (Stx)-producing Escherichia coli (STEC). Most well-known aspects of the pathophysiology are secondary to microthrombotic kidney disease including hemolytic anemia and thrombocytopenia. However, extrarenal manifestations, such as cardiac impairment, have also been reported. We have investigated whether these cardiac abnormalities can be reproduced in a murine animal model, in which administration of Stx, the main virulence factor of STEC, is used to induce HUS. Methods: Mice received either one high or multiple low doses of Stx to simulate the (clinically well-known) different disease courses. Cardiac function was evaluated by echocardiography and analyses of biomarkers in the plasma (troponin I and brain natriuretic peptide). Results: All Stx-challenged mice showed reduced cardiac output and depletion of intravascular volume indicated by a reduced end-diastolic volume and a higher hematocrit. Some mice exhibited myocardial injury (measured as increases in cTNI levels). A subset of mice challenged with either dosage regimen showed hyperkalemia with typical electrocardiographic abnormalities. Discussion: Myocardial injury, intravascular volume depletion, reduced cardiac output, and arrhythmias as a consequence of hyperkalemia may be prognosis-relevant disease manifestations of HUS, the significance of which should be further investigated in future preclinical and clinical studies.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Hyperkalemia , Shiga-Toxigenic Escherichia coli , Animals , Mice , Shiga Toxin 2/toxicity , Pilot Projects , Escherichia coli Infections/complications
2.
Front Immunol ; 14: 1105181, 2023.
Article in English | MEDLINE | ID: mdl-36911665

ABSTRACT

Hemolytic-uremic syndrome (HUS) can occur as a complication of an infection with Shiga-toxin (Stx)-producing Escherichia coli. Patients typically present with acute kidney injury, microangiopathic hemolytic anemia and thrombocytopenia. There is evidence that Stx-induced renal damage propagates a pro-inflammatory response. To date, therapy is limited to organ-supportive strategies. Bruton's tyrosine kinase (BTK) plays a pivotal role in recruitment and function of immune cells and its inhibition was recently shown to improve renal function in experimental sepsis and lupus nephritis. We hypothesized that attenuating the evoked immune response by BTK-inhibitors (BTKi) ameliorates outcome in HUS. We investigated the effect of daily oral administration of the BTKi ibrutinib (30 mg/kg) and acalabrutinib (3 mg/kg) in mice with Stx-induced HUS at day 7. After BTKi administration, we observed attenuated disease progression in mice with HUS. These findings were associated with less BTK and downstream phospholipase-C-gamma-2 activation in the spleen and, subsequently, a reduced renal invasion of BTK-positive cells including neutrophils. Only ibrutinib treatment diminished renal invasion of macrophages, improved acute kidney injury and dysfunction (plasma levels of NGAL and urea) and reduced hemolysis (plasma levels of bilirubin and LDH activity). In conclusion, we report here for the first time that BTK inhibition attenuates the course of disease in murine HUS. We suggest that the observed reduction of renal immune cell invasion contributes - at least in part - to this effect. Further translational studies are needed to evaluate BTK as a potential target for HUS therapy to overcome currently limited treatment options.


Subject(s)
Acute Kidney Injury , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Mice , Animals , Agammaglobulinaemia Tyrosine Kinase , Kidney/physiology , Epithelial Cells , Acute Kidney Injury/complications
3.
Front Immunol ; 13: 1010882, 2022.
Article in English | MEDLINE | ID: mdl-36211426

ABSTRACT

Hemolytic-uremic syndrome (HUS) can occur as a systemic complication of infections with Shiga toxin (Stx)-producing Escherichia coli and is characterized by microangiopathic hemolytic anemia and acute kidney injury. Hitherto, therapy has been limited to organ-supportive strategies. Erythropoietin (EPO) stimulates erythropoiesis and is approved for the treatment of certain forms of anemia, but not for HUS-associated hemolytic anemia. EPO and its non-hematopoietic analog pyroglutamate helix B surface peptide (pHBSP) have been shown to mediate tissue protection via an innate repair receptor (IRR) that is pharmacologically distinct from the erythropoiesis-mediating receptor (EPO-R). Here, we investigated the changes in endogenous EPO levels in patients with HUS and in piglets and mice subjected to preclinical HUS models. We found that endogenous EPO was elevated in plasma of humans, piglets, and mice with HUS, regardless of species and degree of anemia, suggesting that EPO signaling plays a role in HUS pathology. Therefore, we aimed to examine the therapeutic potential of EPO and pHBSP in mice with Stx-induced HUS. Administration of EPO or pHBSP improved 7-day survival and attenuated renal oxidative stress but did not significantly reduce renal dysfunction and injury in the employed model. pHBSP, but not EPO, attenuated renal nitrosative stress and reduced tubular dedifferentiation. In conclusion, targeting the EPO-R/IRR axis reduced mortality and renal oxidative stress in murine HUS without occurrence of thromboembolic complications or other adverse side effects. We therefore suggest that repurposing EPO for the treatment of patients with hemolytic anemia in HUS should be systematically investigated in future clinical trials.


Subject(s)
Erythropoietin , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Animals , Cytokine Receptor Common beta Subunit , Erythropoietin/pharmacology , Hemolytic-Uremic Syndrome/drug therapy , Humans , Mice , Oligopeptides , Receptors, Erythropoietin , Shiga Toxins , Swine
4.
Front Immunol ; 13: 895100, 2022.
Article in English | MEDLINE | ID: mdl-35874776

ABSTRACT

Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.


Subject(s)
Lung , Pneumonia , Air Pollutants , Antigen-Presenting Cells/immunology , Antimicrobial Peptides , Chemokines , Cytokines , Granulocytes/immunology , Humans , Lung/immunology , Mucus/immunology
5.
Kidney Int ; 101(6): 1171-1185, 2022 06.
Article in English | MEDLINE | ID: mdl-35031328

ABSTRACT

Thrombotic microangiopathy, hemolysis and acute kidney injury are typical clinical characteristics of hemolytic-uremic syndrome (HUS), which is predominantly caused by Shiga-toxin-producing Escherichia coli. Free heme aggravates organ damage in life-threatening infections, even with a low degree of systemic hemolysis. Therefore, we hypothesized that the presence of the hemoglobin- and the heme-scavenging proteins, haptoglobin and hemopexin, respectively impacts outcome and kidney pathology in HUS. Here, we investigated the effect of haptoglobin and hemopexin deficiency (haptoglobin-/-, hemopexin-/-) and haptoglobin treatment in a murine model of HUS-like disease. Seven-day survival was decreased in haptoglobin-/- (25%) compared to wild type mice (71.4%), whereas all hemopexin-/- mice survived. Shiga-toxin-challenged hemopexin-/- mice showed decreased kidney inflammation and attenuated thrombotic microangiopathy, indicated by reduced neutrophil recruitment and platelet deposition. These observations were associated with supranormal haptoglobin plasma levels in hemopexin-/- mice. Low dose haptoglobin administration to Shiga-toxin-challenged wild type mice attenuated kidney platelet deposition and neutrophil recruitment, suggesting that haptoglobin at least partially contributes to the beneficial effects. Surrogate parameters of hemolysis were elevated in Shiga-toxin-challenged wild type and haptoglobin-/- mice, while signs for hepatic hemoglobin degradation like heme oxygenase-1, ferritin and CD163 expression were only increased in Shiga-toxin-challenged wild type mice. In line with this observation, haptoglobin-/- mice displayed tubular iron deposition as an indicator for kidney hemoglobin degradation. Thus, haptoglobin and hemopexin deficiency plays divergent roles in Shiga-toxin-mediated HUS, suggesting haptoglobin is involved and hemopexin is redundant for the resolution of HUS pathology.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Thrombotic Microangiopathies , Animals , Disease Progression , Escherichia coli Infections/complications , Haptoglobins/genetics , Heme , Hemoglobins , Hemolysis , Hemolytic-Uremic Syndrome/complications , Hemopexin , Mice , Shiga Toxin , Thrombotic Microangiopathies/etiology
6.
Front Immunol ; 12: 701275, 2021.
Article in English | MEDLINE | ID: mdl-34349763

ABSTRACT

Metabolic endotoxemia has been suggested to play a role in the pathophysiology of metaflammation, insulin-resistance and ultimately type-2 diabetes mellitus (T2DM). The role of endogenous antimicrobial peptides (AMPs), such as the cathelicidin LL-37, in T2DM is unknown. We report here for the first time that patients with T2DM compared to healthy volunteers have elevated plasma levels of LL-37. In a reverse-translational approach, we have investigated the effects of the AMP, peptide 19-2.5, in a murine model of high-fat diet (HFD)-induced insulin-resistance, steatohepatitis and T2DM. HFD-fed mice for 12 weeks caused obesity, an impairment in glycemic regulations, hypercholesterolemia, microalbuminuria and steatohepatitis, all of which were attenuated by Peptide 19-2.5. The liver steatosis caused by feeding mice a HFD resulted in the activation of nuclear factor kappa light chain enhancer of activated B cells (NF-ĸB) (phosphorylation of inhibitor of kappa beta kinase (IKK)α/ß, IκBα, translocation of p65 to the nucleus), expression of NF-ĸB-dependent protein inducible nitric oxide synthase (iNOS) and activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, all of which were reduced by Peptide 19-2.5. Feeding mice, a HFD also resulted in an enhanced expression of the lipid scavenger receptor cluster of differentiation 36 (CD36) secondary to activation of extracellular signal-regulated kinases (ERK)1/2, both of which were abolished by Peptide 19-2.5. Taken together, these results demonstrate that the AMP, Peptide 19-2.5 reduces insulin-resistance, steatohepatitis and proteinuria. These effects are, at least in part, due to prevention of the expression of CD36 and may provide further evidence for a role of metabolic endotoxemia in the pathogenesis of metaflammation and ultimately T2DM. The observed increase in the levels of the endogenous AMP LL-37 in patients with T2DM may serve to limit the severity of the disease.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Antimicrobial Peptides/pharmacology , Diabetes Mellitus, Type 2/metabolism , Inflammation , Lipopolysaccharides/antagonists & inhibitors , Animals , Diet, High-Fat/adverse effects , Endotoxemia/etiology , Endotoxemia/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Cathelicidins
7.
Front Immunol ; 11: 581758, 2020.
Article in English | MEDLINE | ID: mdl-33162995

ABSTRACT

We previously reported the Bruton's tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib improve outcomes in a mouse model of polymicrobial sepsis. Now we show that genetic deficiency of the BTK gene alone in Xid mice confers protection against cardiac, renal, and liver injury in polymicrobial sepsis and reduces hyperimmune stimulation ("cytokine storm") induced by an overwhelming bacterial infection. Protection is due in part to enhanced bacterial phagocytosis in vivo, changes in lipid metabolism and decreased activation of NF-κB and the NLRP3 inflammasome. The inactivation of BTK leads to reduced innate immune cell recruitment and a phenotypic switch from M1 to M2 macrophages, aiding in the resolution of sepsis. We have also found that BTK expression in humans is increased in the blood of septic non-survivors, while lower expression is associated with survival from sepsis. Importantly no further reduction in organ damage, cytokine production, or changes in plasma metabolites is seen in Xid mice treated with the BTK inhibitor ibrutinib, demonstrating that the protective effects of BTK inhibitors in polymicrobial sepsis are mediated solely by inhibition of BTK and not by off-target effects of this class of drugs.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Multiple Organ Failure/metabolism , Sepsis/metabolism , X-Linked Combined Immunodeficiency Diseases/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Benzamides/pharmacology , Disease Models, Animal , Inflammasomes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred CBA , Multiple Organ Failure/drug therapy , Phagocytosis/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Sepsis/drug therapy , X-Linked Combined Immunodeficiency Diseases/drug therapy
8.
JCI Insight ; 5(8)2020 04 23.
Article in English | MEDLINE | ID: mdl-32213712

ABSTRACT

Septic cardiomyopathy is a life-threatening organ dysfunction caused by sepsis. Ribonuclease 1 (RNase 1) belongs to a group of host-defense peptides that specifically cleave extracellular RNA (eRNA). The activity of RNase 1 is inhibited by ribonuclease-inhibitor 1 (RNH1). However, the role of RNase 1 in septic cardiomyopathy and associated cardiac apoptosis is completely unknown. Here, we show that sepsis resulted in a significant increase in RNH1 and eRNA serum levels compared with those of healthy subjects. Treatment with RNase 1 resulted in a significant decrease of apoptosis, induced by the intrinsic pathway, and TNF expression in murine cardiomyocytes exposed to either necrotic cardiomyocytes or serum of septic patients for 16 hours. Additionally, treatment of septic mice with RNase 1 resulted in a reduction in cardiac apoptosis, TNF expression, and septic cardiomyopathy. These data demonstrate that eRNA plays a crucial role in the pathophysiology of the organ (cardiac) dysfunction in sepsis and that RNase and RNH1 may be new therapeutic targets and/or strategies to reduce the cardiac injury and dysfunction caused by sepsis.


Subject(s)
Cardiomyopathies/metabolism , Cell-Free Nucleic Acids/metabolism , Ribonuclease, Pancreatic/metabolism , Sepsis/metabolism , Animals , Apoptosis/physiology , Cardiomyopathies/etiology , Carrier Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Proteins/metabolism , Sepsis/complications
9.
Front Immunol ; 10: 2129, 2019.
Article in English | MEDLINE | ID: mdl-31552054

ABSTRACT

Sepsis is one of the most prevalent diseases in the world. The development of cardiac dysfunction in sepsis results in an increase of mortality. It is known that Bruton's tyrosine kinase (BTK) plays a role in toll-like receptor signaling and NLRP3 inflammasome activation, two key components in the pathophysiology of sepsis and sepsis-associated cardiac dysfunction. In this study we investigated whether pharmacological inhibition of BTK (ibrutinib 30 mg/kg and acalabrutinib 3 mg/kg) attenuates sepsis associated cardiac dysfunction in mice. 10-week old male C57BL/6 mice underwent CLP or sham surgery. One hour after surgery mice received either vehicle (5% DMSO + 30% cyclodextrin i.v.), ibrutinib (30 mg/kg i.v.), or acalabrutinib (3 mg/kg i.v.). Mice also received antibiotics and an analgesic at 6 and 18 h. After 24 h, cardiac function was assessed by echocardiography in vivo. Cardiac tissue underwent western blot analysis to determine the activation of BTK, NLRP3 inflammasome and NF-κB pathway. Serum analysis of 33 cytokines was conducted by a multiplex assay. When compared to sham-operated animals, mice subjected to CLP demonstrated a significant reduction in ejection fraction (EF), fractional shortening (FS), and fractional area change (FAC). The cardiac tissue from CLP mice showed significant increases of BTK, NF-κB, and NLRP3 inflammasome activation. CLP animals resulted in a significant increase of serum cytokines and chemokines (TNF-α, IL-6, IFN-γ, KC, eotaxin-1, eotaxin-2, IL-10, IL-4, CXCL10, and CXCL11). Delayed administration of ibrutinib and acalabrutinib attenuated the decline of EF, FS, and FAC caused by CLP and also reduced the activation of BTK, NF-κB, and NLRP3 inflammasome. Both ibrutinib and acalabrutinib significantly suppressed the release of cytokines and chemokines. Our study revealed that delayed intravenous administration of ibrutinib or acalabrutinib attenuated the cardiac dysfunction associated with sepsis by inhibiting BTK, reducing NF-κB activation and the activation of the inflammasome. Cytokines associated with sepsis were significantly reduced by both BTK inhibitors. Acalabrutinib is found to be more potent than ibrutinib and could potentially prove to be a novel therapeutic in sepsis. Thus, the FDA-approved BTK inhibitors ibrutinib and acalabrutinib may be repurposed for the use in sepsis.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Heart Diseases/etiology , Heart/drug effects , Protein Kinase Inhibitors/pharmacology , Sepsis/complications , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/drug effects , Agammaglobulinaemia Tyrosine Kinase/immunology , Animals , Benzamides/pharmacology , Cecum , Disease Models, Animal , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Ligation , Male , Mice , Mice, Inbred C57BL , Piperidines , Punctures , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sepsis/immunology , Sepsis/metabolism
10.
Front Immunol ; 9: 1459, 2018.
Article in English | MEDLINE | ID: mdl-29988557

ABSTRACT

Diarrhea-positive hemolytic-uremic syndrome (HUS) is a renal disorder that results from infections with Shiga-toxin (Stx)-producing Escherichia coli. The aim of this study was to establish well-defined refined murine models of HUS that can serve as preclinical tools to elucidate molecular mechanisms of disease development. C57BL/6J mice were subjected to different doses of Stx2 purified from an E. coli O157:H7 patient isolate. Animals received 300 ng/kg Stx2 and were sacrificed on day 3 to establish an acute model with fast disease progression. Alternatively, mice received 25 ng/kg Stx2 on days 0, 3, and 6, and were sacrificed on day 7 to establish a subacute model with moderate disease progression. Indicated by a rise in hematocrit, we observed dehydration despite volume substitution in both models, which was less pronounced in mice that underwent the 7-day regime. Compared with sham-treated animals, mice subjected to Stx2 developed profound weight loss, kidney dysfunction (elevation of plasma urea, creatinine, and neutrophil gelatinase-associated lipocalin), kidney injury (tubular injury and loss of endothelial cells), thrombotic microangiopathy (arteriolar microthrombi), and hemolysis (elevation of plasma bilirubin, lactate dehydrogenase, and free hemoglobin). The degree of complement activation (C3c deposition), immune cell invasion (macrophages and T lymphocytes), apoptosis, and proliferation were significantly increased in kidneys of mice subjected to the 7-day but not in kidneys of mice subjected to the 3-day regime. However, glomerular and kidney volume remained mainly unchanged, as assessed by 3D analysis of whole mount kidneys using CD31 staining with light sheet fluorescence microscopy. Gene expression analysis of kidneys revealed a total of only 91 overlapping genes altered in both Stx2 models. In conclusion, we have developed two refined mouse models with different disease progression, both leading to hemolysis, thrombotic microangiopathy, and acute kidney dysfunction and damage as key clinical features of human HUS. While intrarenal changes (apoptosis, proliferation, complement deposition, and immune cell invasion) mainly contribute to the pathophysiology of the subacute model, prerenal pathomechanisms (hypovolemia) play a predominant role in the acute model. Both models allow the further study of the pathomechanisms of most aspects of human HUS and the testing of distinct novel treatment strategies.

11.
Life Sci ; 203: 112-120, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29684444

ABSTRACT

AIMS: Studies on omega-3 fatty acids, including docosahexaenoic acid (DHA), reveal diverging results: Their intake is recommended in cardiovascular disease and major surgery, while evidence argues against use in septic patients. DHA mediates its blood-pressure-lowering effect through Slo1 channels that are expressed on cardiovascular and immune cells. We hypothesised that conflicting effects of immunonutrition could be explained by the influence of omega-3 fatty acids on systemic blood pressure or immune effector cells through Slo1. MAIN METHODS: The effect of DHA on blood pressure was analysed in septic wild-type (WT) mice. Septic WT and Slo1 knockout (KO) mice were compared regarding survival, clinical presentation, haematology, cytokine release and bacterial burden. Cytokine expression and release of bone marrow derived macrophages (BMDM) from WT and Slo1 KO mice was assessed in response to LPS. KEY FINDINGS: The significant blood-pressure-lowering effect of DHA in healthy animals was blunted in already hypotensive septic mice. Septic Slo1 KO mice displayed moderately lower bacterial burden in blood and lungs compared with WT, which did not translate into improved survival. Slo1 KO BMDM presented lower IL-6 levels in response to LPS, an effect that was abolished in the presence of DHA. More importantly, the strong inhibitory effect of DHA on IL-6 release was also observed in Slo1 KO BMDM. SIGNIFICANCE: The controversial effects of immunonutrition in sepsis are unlikely to be primarily explained by the influence of DHA on blood pressure or effects on immune response mediated through Slo1 channels.


Subject(s)
Docosahexaenoic Acids/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Lung/immunology , Macrophages/immunology , Sepsis/immunology , Animals , Blood Pressure/drug effects , Cells, Cultured , Cytokines/metabolism , Female , Lung/metabolism , Lung/microbiology , Lung/pathology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mice, Knockout , Sepsis/metabolism , Sepsis/pathology
12.
Pharmacol Res ; 87: 80-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24973659

ABSTRACT

In the present study, the neuroprotective effects of the adipokine leptin, and the molecular mechanism involved, have been studied in rat and mice cortical neurons exposed to N-methyl-d-aspartate (NMDA) in vitro. In rat cortical neurons, leptin elicited neuroprotective effects against NMDA-induced cell death, which were concentration-dependent (10-100 ng/ml) and largest when the adipokine was preincubated for 2h before the neurotoxic stimulus. In both rat and mouse cortical neurons, leptin-induced neuroprotection was fully antagonized by paxilline (Pax, 0.01-1 µM) and iberiotoxin (Ibtx, 1-100 nM), with EC50s of 38 ± 10 nM and 5 ± 2 nM for Pax and Ibtx, respectively, close to those reported for Pax- and Ibtx-induced Ca(2+)- and voltage-activated K(+) channels (Slo1 BK channels) blockade; the BK channel opener NS1619 (1-30 µM) induced a concentration-dependent protection against NMDA-induced excitotoxicity. Moreover, cortical neurons from mice lacking one or both alleles coding for Slo1 BK channel pore-forming subunits were insensitive to leptin-induced neuroprotection. Finally, leptin exposure dose-dependently (10-100 ng/ml) increased intracellular Ca(2+) levels in rat cortical neurons. In conclusion, our results suggest that Slo1 BK channel activation following increases in intracellular Ca(2+) levels is a critical step for leptin-induced neuroprotection in NMDA-exposed cortical neurons in vitro, thus highlighting leptin-based intervention via BK channel activation as a potential strategy to counteract neurodegenerative diseases.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Leptin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Mice, Transgenic , N-Methylaspartate , Neurons/metabolism , Rats, Wistar
13.
PLoS One ; 8(10): e77224, 2013.
Article in English | MEDLINE | ID: mdl-24098580

ABSTRACT

The single nucleotide polymorphism rs2071746 and a (GT)n microsatellite within the human gene encoding heme oxygenase-1 (HMOX1) are associated with incidence or outcome in a variety of diseases. Most of these associations involve either release of heme or oxidative stress. Both polymorphisms are localized in the promoter region, but previously reported correlations with heme oxygenase-1 expression remain not coherent. This ambiguity suggests a more complex organization of the 5' gene region which we sought to investigate more fully. We evaluated the 5' end of HMOX1 and found a novel first exon 1a placing the two previously reported polymorphisms in intronic or exonic positions within the 5' untranslated region respectively. Expression of exon 1a can be induced in HepG2 hepatoma cells by hemin and is a repressor of heme oxygenase-1 translation as shown by luciferase reporter assays. Moreover, minigene approaches revealed that the quantitative outcome of alternative splicing within the 5' untranslated region is affected by the (GT)n microsatellite. This data supporting an extended HMOX1 gene model and provide further insights into expression regulation of heme oxygenase-1. Alternative splicing within the HMOX1 5' untranslated region contributes to translational regulation and is a mechanistic feature involved in the interplay between genetic variations, heme oxygenase-1 expression and disease outcome.


Subject(s)
5' Untranslated Regions , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Alternative Splicing , Base Sequence , Exons , Genes, Reporter , Heme Oxygenase-1/metabolism , Hemin/pharmacology , Hep G2 Cells , Humans , Introns , Luciferases/genetics , Luciferases/metabolism , Microsatellite Repeats , Molecular Sequence Data
15.
Crit Care Med ; 41(10): e246-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23887230

ABSTRACT

OBJECTIVES: High physical activity levels are associated with wide-ranging health benefits, disease prevention, and longevity. In the present study, we examined the impact of regular physical exercise on the severity of organ injury and survival probability, as well as characteristics of the systemic immune and metabolic response during severe polymicrobial sepsis. DESIGN: Animal study. SETTING: University laboratory. SUBJECTS: Male C57BL/6N mice. INTERVENTIONS: Mice were trained for 6 weeks by treadmill and voluntary wheel running or housed normally. Polymicrobial sepsis in mice was induced by injection of fecal slurry. Subsequently, mice were randomized into the following groups: healthy controls, 6 hours postsepsis, and 24 hours postsepsis. MEASUREMENTS AND MAIN RESULTS: Blood and organ samples were collected and investigated by measuring clinical chemistry variables, cytokines, plasma metabolites, and bacterial clearance. Organ morphology and damage were characterized by histological staining. Physical exercise improved survival and the ability of bacterial clearance in blood and organs. The release of pro- and anti-inflammatory cytokines, including interleukin-6 and interleukin-10, was diminished in trained compared to untrained mice during sepsis. The sepsis-associated acute kidney tubular damage was less pronounced in pretrained animals. By metabolic profiling and regression analysis, we detected lysophosphatidylcholine 14:0, tryptophan, as well as pimelylcarnitine linked with levels of neutrophil gelatinase-associated lipocalin representing acute tubular injury (corrected R=0.910; p<0.001). We identified plasma lysophosphatidylcholine 16:0, lysophosphatidylcholine 17:0, and lysophosphatidylcholine 18:0 as significant metabolites discriminating between trained and untrained mice during sepsis. CONCLUSIONS: Regular physical exercise reduces sepsis-associated acute kidney injury and death. As a specific mechanism of exercise-induced adaptation, we identified various lysophosphatidylcholines that might function as surrogate for improved outcome in sepsis.


Subject(s)
Acute Kidney Injury/prevention & control , Coinfection/complications , Hepatic Insufficiency/prevention & control , Lung Injury/prevention & control , Physical Conditioning, Animal , Sepsis/complications , Acute Kidney Injury/metabolism , Acute Kidney Injury/microbiology , Adaptation, Physiological/immunology , Animals , Coinfection/mortality , Cytokines/metabolism , Hepatic Insufficiency/metabolism , Hepatic Insufficiency/microbiology , Lung Injury/metabolism , Lung Injury/microbiology , Male , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal/methods , Random Allocation , Sepsis/mortality , Survival Analysis
16.
Proc Natl Acad Sci U S A ; 110(12): 4816-21, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23487785

ABSTRACT

Long-chain polyunsaturated omega-3 fatty acids such as docosahexaenoic acid (DHA), found abundantly in oily fish, may have diverse health-promoting effects, potentially protecting the immune, nervous, and cardiovascular systems. However, the mechanisms underlying the purported health-promoting effects of DHA remain largely unclear, in part because molecular signaling pathways and effectors of DHA are only beginning to be revealed. In vascular smooth muscle cells, large-conductance Ca(2+)- and voltage-activated K(+) (BK) channels provide a critical vasodilatory influence. We report here that DHA with an EC50 of ∼500 nM rapidly and reversibly activates BK channels composed of the pore-forming Slo1 subunit and the auxiliary subunit ß1, increasing currents by up to ∼20-fold. The DHA action is observed in cell-free patches and does not require voltage-sensor activation or Ca(2+) binding but involves destabilization of the closed conformation of the ion conduction gate. DHA lowers blood pressure in anesthetized wild-type but not in Slo1 knockout mice. DHA ethyl ester, contained in dietary supplements, fails to activate BK channels and antagonizes the stimulatory effect of DHA. Slo1 BK channels are thus receptors for long-chain omega-3 fatty acids, and these fatty acids--unlike their ethyl ester derivatives--activate the channels and lower blood pressure. This finding has practical implications for the use of omega-3 fatty acids as nutraceuticals for the general public and also for the critically ill receiving omega-3-enriched formulas.


Subject(s)
Blood Pressure/drug effects , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Vasodilation/drug effects , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/drug effects , Mice , Mice, Knockout , Protein Structure, Tertiary
18.
Free Radic Biol Med ; 51(2): 432-43, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21570463

ABSTRACT

Ascorbic acid (AA) increases cardiomyogenesis of embryonic stem (ES) cells. Herein we show that treatment of mouse ES cells with AA enhanced cardiac differentiation accompanied by an upregulation of the NADPH oxidase isoforms NOX2 and NOX4, phosphorylation of endothelial nitric oxide synthase (eNOS), and cyclic GMP (cGMP) formation, indicating that reactive oxygen species (ROS) as well as nitric oxide (NO) may be involved in cardiomyogenesis. In whole mount embryoid bodies as well as isolated Flk-1-positive (Flk-1(+)) cardiovascular progenitor cells ROS elevation by AA was observed in early stages of differentiation (Days 4-7), and absent at Day 10. In contrast NO generation following incubation with AA was absent at Day 4 and increased at Days 7 and 10. AA-mediated cardiomyogenesis was blunted by the NADPH oxidase inhibitors diphenylen iodonium (DPI) and apocynin, the free radical scavengers N-(2-mercaptopropionyl)-glycine (NMPG) and ebselen, and the NOS inhibitor L-NAME. Downregulation of NOX4 by short hairpin RNA (shRNA) resulted in significant inhibition of cardiomyogenesis and abolished the stimulation of MHC-ß and MLC2v gene expression observed on AA treatment. Our data demonstrate that AA stimulates cardiomyocyte differentiation from ES cells by signaling pathways that involve ROS generated at early stages and NO at late stages of cardiomyogenesis.


Subject(s)
Ascorbic Acid/pharmacology , Embryonic Stem Cells/drug effects , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Base Sequence , Blotting, Western , Cell Differentiation , Cell Lineage , DNA Primers , Embryonic Stem Cells/cytology , Mice , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
J Agric Food Chem ; 56(17): 7649-54, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18698790

ABSTRACT

The exact and reliable determination of carbohydrates in plant samples of different origin is of great importance with respect to plant physiology. Additionally, the identification and quantification of carbohydrates are necessary for the evaluation of the impact of these compounds on the biogeochemistry of carbon. To attain this goal, it is necessary to analyze a great number of samples with both high sensitivity and selectivity within a limited time frame. This paper presents a rugged and easy method that allows the isocratic chromatographic determination of 12 carbohydrates and sugar alcohols from one sample within 30 min. The method was successfully applied to a variety of plant materials with particular emphasis on perennial ryegrass samples of the species Lolium perenne. The method was easily extended to the analysis of the polysaccharide inulin after its acidic hydrolysis into the corresponding monomers without the need for substantial change of chromatographic conditions or even the use of enzymes. It therefore offers a fundamental advantage for the analysis of the complex mixture of nonstructural carbohydrates often found in plant samples.


Subject(s)
Carbohydrates/analysis , Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Inulin/analysis , Poaceae/chemistry , Lolium/chemistry , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...