Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Clin Kidney J ; 16(11): 1786-1803, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37915935

RÉSUMÉ

The cardiorenal syndrome (CRS) is described as a multi-organ disease encompassing bidirectionally heart and kidney. In CRS type 4, chronic kidney disease (CKD) leads to cardiac injury. Different pathological mechanisms have been identified to contribute to the establishment of CKD-induced cardiomyopathy, including a neurohormonal dysregulation, disturbances in the mineral metabolism and an accumulation of uremic toxins, playing an important role in the development of inflammation and oxidative stress. Combined, this leads to cardiac dysfunction and cardiac pathophysiological and morphological changes, like left ventricular hypertrophy, myocardial fibrosis and cardiac electrical changes. Given that around 80% of dialysis patients suffer from uremic cardiomyopathy, the study of cardiac outcomes in CKD is clinically highly relevant. The present review summarizes clinical features and biomarkers of CKD-induced cardiomyopathy and discusses underlying pathophysiological mechanisms recently uncovered in the literature. It discloses how animal models have contributed to the understanding of pathological kidney-heart crosstalk, but also provides insights into the variability in observed effects of CKD on the heart in different CKD mouse models, covering both "single hit" as well as "multifactorial hit" models. Overall, this review aims to support research progress in the field of CKD-induced cardiomyopathy.

2.
Kidney Int ; 103(6): 1199-1201, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37210195
3.
Clin Gastroenterol Hepatol ; 21(11): 2746-2758, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-36470528

RÉSUMÉ

BACKGROUND & AIMS: Growing evidence supports a role of gut-derived metabolites in nonalcoholic fatty liver disease (NAFLD), but the relation of endotoxin levels with gut permeability and NAFLD stage remains unclear. This systematic review with meta-analysis aims to provide further insights. METHODS: PubMed, Embase, and Cochrane Library were searched for studies published until January 2022 assessing blood endotoxins in patients with NAFLD. Meta-analyses and univariate/multivariate meta-regression, as well as correlation analyses, were performed for endotoxin values and potential relationships to disease stage, age, sex, parameters of systemic inflammation, and metabolic syndrome, as well as liver function and histology. RESULTS: Forty-three studies were included, of which 34 were used for meta-analyses. Blood endotoxin levels were higher in patients with simple steatosis vs liver-healthy controls (standardized mean difference, 0.86; 95% confidence interval, 0.62-1.11) as well as in patients with nonalcoholic steatohepatitis vs patients with nonalcoholic fatty liver/non-nonalcoholic steatohepatitis (standardized mean difference, 0.81; 95% confidence interval, 0.27-1.35; P = .0078). Consistently, higher endotoxin levels were observed in patients with more advanced histopathological gradings of liver steatosis and fibrosis. An increase of blood endotoxin levels was partially attributed to a body mass index rise in patients with NAFLD compared with controls. Nevertheless, significant increases of blood endotoxin levels in NAFLD retained after compensation for differences in body mass index, metabolic condition, or liver enzymes. Increases in blood endotoxin levels were associated with increases in C-reactive protein concentrations, and in most cases, paralleled a rise in markers for intestinal permeability. CONCLUSION: Our results support blood endotoxin levels as relevant diagnostic biomarker for NAFLD, both for disease detection as well as staging during disease progression, and might serve as surrogate marker of enhanced intestinal permeability in NAFLD. Registration number in Prospero: CRD42022311166.


Sujet(s)
Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/anatomopathologie , Endotoxines/métabolisme , Foie/anatomopathologie , Inflammation/anatomopathologie , Marqueurs biologiques/métabolisme
4.
Kidney Int ; 101(2): 256-273, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34774555

RÉSUMÉ

Chronic kidney disease (CKD) triggers the risk of developing uremic cardiomyopathy as characterized by cardiac hypertrophy, fibrosis and functional impairment. Traditionally, animal studies are used to reveal the underlying pathological mechanism, although variable CKD models, mouse strains and readouts may reveal diverse results. Here, we systematically reviewed 88 studies and performed meta-analyses of 52 to support finding suitable animal models for future experimental studies on pathological kidney-heart crosstalk during uremic cardiomyopathy. We compared different mouse strains and the direct effect of CKD on cardiac hypertrophy, fibrosis and cardiac function in "single hit" strategies as well as cardiac effects of kidney injury combined with additional cardiovascular risk factors in "multifactorial hit" strategies. In C57BL/6 mice, CKD was associated with a mild increase in cardiac hypertrophy and fibrosis and marginal systolic dysfunction. Studies revealed high variability in results, especially regarding hypertrophy and systolic function. Cardiac hypertrophy in CKD was more consistently observed in 129/Sv mice, which express two instead of one renin gene and more consistently develop increased blood pressure upon CKD induction. Overall, "multifactorial hit" models more consistently induced cardiac hypertrophy and fibrosis compared to "single hit" kidney injury models. Thus, genetic factors and additional cardiovascular risk factors can "prime" for susceptibility to organ damage, with increased blood pressure, cardiac hypertrophy and early cardiac fibrosis more consistently observed in 129/Sv compared to C57BL/6 strains.


Sujet(s)
Cardiomyopathies , Insuffisance rénale chronique , Animaux , Cardiomyopathies/génétique , Modèles animaux de maladie humaine , Fibrose , Souris , Souris de lignée C57BL , Insuffisance rénale chronique/complications
5.
Front Physiol ; 12: 638448, 2021.
Article de Anglais | MEDLINE | ID: mdl-34366876

RÉSUMÉ

Background: Adipose-derived stem cells (ASCs) are multipotent mesenchymal stem cells characterized by their strong regenerative potential and low oxygen consumption. Macrophage migration inhibitory factor (MIF) is a multifunctional chemokine-like cytokine that is involved in tissue hypoxia. MIF is not only a major immunomodulator but also is highly expressed in adipose tissue such as subcutaneous adipose tissue of chronic non-healing wounds. In the present study, we investigated the effect of hypoxia on MIF in ASCs isolated from healthy versus inflamed adipose tissue. Methods: Human ASCs were harvested from 17 patients (11 healthy adipose tissue samples, six specimens from chronic non-healing wounds). ASCs were treated in a hypoxia chamber at <1% oxygen. ASC viability, MIF secretion as well as expression levels of MIF, its receptor CD74, hypoxia-inducible transcription factor-1α (HIF-1α) and activation of the AKT and ERK signaling pathways were analyzed. The effect of recombinant MIF on the viability of ASCs was determined. Finally, the effect of MIF on the viability and production capacity of ASCs to produce the inflammatory cytokines tumor necrosis factor (TNF), interleukin (IL)-6, and IL-1ß was determined upon treatment with recombinant MIF and/or a blocking MIF antibody. Results: Hypoxic treatment inhibited proliferation of ASCs derived from healthy or chronic non-healing wounds. ASCs from healthy adipose tissue samples were characterized by a low degree of MIF secretion during hypoxic challenge. In contrast, in ASCs from adipose tissue samples of chronic non-healing wounds, secretion and expression of MIF and CD74 expression were significantly elevated under hypoxia. This was accompanied by enhanced ERK signaling, while AKT signaling was not altered. Recombinant MIF did stimulate HIF-1α expression under hypoxia as well as AKT and ERK phosphorylation, while no effect on ASC viability was observed. Recombinant MIF significantly reduced the secretion of IL-1ß under hypoxia and normoxia, and neutralizing MIF-antibodies diminished TNF-α and IL-1ß release in hypoxic ASCs. Conclusions: Collectively, MIF did not affect the viability of ASCs from neither healthy donor site nor chronic wounds. Our results, however, suggest that MIF has an impact on the wound environment by modulating inflammatory factors such as IL-1ß.

6.
Int J Mol Sci ; 22(9)2021 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-33922385

RÉSUMÉ

Phosphatidylserines are known to sustain skeletal muscle activity during intense activity or hypoxic conditions, as well as preserve neurocognitive function in older patients. Our previous studies pointed out a potential cardioprotective role of phosphatidylserine in heart ischemia. Therefore, we investigated the effects of phosphatidylserine oral supplementation in a mouse model of acute myocardial infarction (AMI). We found out that phosphatidylserine increases, significantly, the cardiomyocyte survival by 50% in an acute model of myocardial ischemia-reperfusion. Similar, phosphatidylserine reduced significantly the infarcted size by 30% and improved heart function by 25% in a chronic model of AMI. The main responsible mechanism seems to be up-regulation of protein kinase C epsilon (PKC-ε), the main player of cardio-protection during pre-conditioning. Interestingly, if the phosphatidylserine supplementation is started before induction of AMI, but not after, it selectively inhibits neutrophil's activation, such as Interleukin 1 beta (IL-1ß) expression, without affecting the healing and fibrosis. Thus, phosphatidylserine supplementation may represent a simple way to activate a pre-conditioning mechanism and may be a promising novel strategy to reduce infarct size following AMI and to prevent myocardial injury during myocardial infarction or cardiac surgery. Due to the minimal adverse effects, further investigation in large animals or in human are soon possible to establish the exact role of phosphatidylserine in cardiac diseases.


Sujet(s)
Compléments alimentaires , Infarctus du myocarde/prévention et contrôle , Lésion de reperfusion myocardique/prévention et contrôle , Phosphatidylsérine/pharmacologie , Dysfonction ventriculaire gauche/complications , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Animaux , Animaux nouveau-nés , Mâle , Souris , Souris de lignée C57BL , Infarctus du myocarde/étiologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Lésion de reperfusion myocardique/étiologie , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Dysfonction ventriculaire gauche/physiopathologie , Remodelage ventriculaire/physiologie
7.
Diagnostics (Basel) ; 11(2)2021 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-33671433

RÉSUMÉ

Mortality in critically ill coronavirus disease 2019 (COVID-19) patients is high and pharmacological treatment strategies remain limited. Early-stage predictive biomarkers are needed to identify patients with a high risk of severe clinical courses and to stratify treatment strategies. Macrophage migration inhibitory factor (MIF) was previously described as a potential predictor for the outcome of critically ill patients and for acute respiratory distress syndrome (ARDS), a hallmark of severe COVID-19 disease. This prospective observational study evaluates the predictive potential of MIF for the clinical outcome after severe COVID-19 infection. Plasma MIF concentrations were measured in 36 mechanically ventilated COVID-19 patients over three days after intensive care unit (ICU) admission. Increased compared to decreased MIF was significantly associated with aggravated organ function and a significantly lower 28-day survival (sequential organ failure assessment (SOFA) score; 8.2 ± 4.5 to 14.3 ± 3, p = 0.009 vs. 8.9 ± 1.9 to 12 ± 2, p = 0.296; survival: 56% vs. 93%; p = 0.003). Arterial hypertension was the predominant comorbidity in 85% of patients with increasing MIF concentrations (vs. decreasing MIF: 39%; p = 0.015). Without reaching significance, more patients with decreasing MIF were able to improve their ARDS status (p = 0.142). The identified association between an early MIF response, aggravation of organ function and 28-day survival may open future perspectives for biomarker-based diagnostic approaches for ICU management of COVID-19 patients.

8.
Adv Drug Deliv Rev ; 159: 4-33, 2020.
Article de Anglais | MEDLINE | ID: mdl-32730849

RÉSUMÉ

With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.


Sujet(s)
Maladies cardiovasculaires/métabolisme , Métabolisme lipidique , Animaux , Maladies cardiovasculaires/prévention et contrôle , Maladies cardiovasculaires/thérapie , Humains , Lipides
9.
Atherosclerosis ; 292: 23-30, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31733453

RÉSUMÉ

BACKGROUND AND AIMS: IKKα is an important regulator of gene expression. As IKKα kinase inactivity in bone marrow-derived cells does not affect atherosclerosis, we here investigate the impact of a whole body-IKKα kinase inactivity on atherosclerosis. METHODS: Apolipoprotein E (Apoe)-deficient mice homozygous for an activation-resistant Ikkα-mutant (IkkαAA/AAApoe-/-) and Ikkα+/+Apoe-/- controls received a Western-type diet. Atherosclerotic lesion size and cellular content were analyzed using histology and immunofluorescence. Vascular protein expression and IKKα kinase activity were quantified by Luminex multiplex immuno-assay and ELISA. RESULTS: A vascular site-specific IKKα expression and kinase activation profile was revealed, with higher total IKKα protein levels in aortic root but increased IKKα phosphorylation, representing activated IKKα, in the aortic arch. This was associated with a vascular site-specific effect of IkkαAA/AA knock-in on atherosclerosis: in the aortic root, IkkαAA/AA knock-in decreased lesion size by 22.0 ±â€¯7.7% (p < 0.01), reduced absolute lesional smooth muscle cell numbers and lowered pro-atherogenic MMP2. In contrast, IkkαAA/AA knock-in increased lesion size in the aortic arch by 43.7 ±â€¯20.1% (p < 0.001), increased the abundance of lesional smooth muscle cells in brachiocephalic artery as main arch side branch and elevated MMP2. A similar profile was observed for MMP3. No effects were observed on necrotic core or collagen deposition in atherosclerotic lesions, nor on absolute lesional macrophage numbers. CONCLUSIONS: A non-activatable IKKα kinase differentially affects atherosclerosis in aortic root vs. aortic arch/brachiocephalic artery, associated with a differential vascular IKKα expression and kinase activation profile as well as with a vascular site-dependent impact on lesional smooth muscle cell accumulation and protein expression profiles.


Sujet(s)
Athérosclérose/étiologie , I-kappa B Kinase/physiologie , Animaux , Apolipoprotéines E/déficit , Athérosclérose/métabolisme , I-kappa B Kinase/antagonistes et inhibiteurs , I-kappa B Kinase/génétique , Souris , Mutation
10.
J Am Heart Assoc ; 7(17): e009384, 2018 09 04.
Article de Anglais | MEDLINE | ID: mdl-30371153

RÉSUMÉ

Background Although macrophage migration inhibitory factor ( MIF ) has been demonstrated to mediate cardioprotection in ischemia/reperfusion injury and antagonize fibrotic effects through its receptor, CD 74, the function of the soluble CD 74 receptor ectodomain ( sCD 74) and its interaction with circulating MIF have not been explored in cardiac disease. Methods and Results Cardiac fibroblasts were isolated from hearts of neonatal mice and differentiated into myofibroblasts. Co-treatment with recombinant MIF and sCD 74 induced cell death ( P<0.001), which was mediated by receptor-interacting serine/threonine-protein kinase ( RIP) 1/ RIP 3-dependent necroptosis ( P=0.0376). This effect was specific for cardiac fibroblasts and did not affect cardiomyocytes. Gene expression analyses using microarray and RT - qPCR technology revealed a 4-fold upregulation of several interferon-induced genes upon co-treatment of myofibroblasts with sCD 74 and MIF (Ifi44: P=0.011; Irg1: P=0.022; Clec4e: P=0.011). Furthermore, Western blot analysis confirmed the role of sCD 74 as a modulator of MIF signaling by diminishing MIF -mediated protein kinase B ( AKT) activation ( P=0.0197) and triggering p38 activation ( P=0.0641). We obtained evidence that sCD 74 inhibits MIF -mediated survival pathway through the C-X-C chemokine receptor 4/ AKT axis, enabling the induction of CD 74-dependent necroptotic processes in cardiac myofibroblasts. Preliminary clinical data revealed a lowered sCD 74/ MIF ratio in heart failure patients (17.47±10.09 versus 1.413±0.6244). Conclusions These findings suggest that treatment of cardiac myofibroblasts with sCD 74 and MIF induces necroptosis, offering new insights into the mechanism of myofibroblast depletion during scar maturation. Preliminary clinical data provided first evidence about a clinical relevance of the sCD 74/ MIF axis in heart failure, suggesting that these proteins may be a promising target to modulate cardiac remodeling and disease progression in heart failure.


Sujet(s)
Antigènes de différenciation des lymphocytes B/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Antigènes d'histocompatibilité de classe II/pharmacologie , Facteurs inhibiteurs de la migration des macrophages/pharmacologie , Myofibroblastes/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs CXCR4/métabolisme , Animaux , Antigènes de différenciation des lymphocytes B/métabolisme , Survie cellulaire , Maladie coronarienne/métabolisme , Protéines d'activation de la GTPase/métabolisme , Expression des gènes , Défaillance cardiaque/métabolisme , Antigènes d'histocompatibilité de classe II/métabolisme , Humains , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Souris , Myocarde , Myocytes cardiaques/effets des médicaments et des substances chimiques , Nécrose , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme
11.
Sci Transl Med ; 10(441)2018 05 16.
Article de Anglais | MEDLINE | ID: mdl-29769287

RÉSUMÉ

Acute kidney injury (AKI) represents the most frequent complication after cardiac surgery. Macrophage migration inhibitory factor (MIF) is a stress-regulating cytokine that was shown to protect the heart from myocardial ischemia-reperfusion injury, but its role in the pathogenesis of AKI remains unknown. In an observational study, serum and urinary MIF was quantified in 60 patients scheduled for elective conventional cardiac surgery with the use of cardiopulmonary bypass. Cardiac surgery triggered an increase in MIF serum concentrations, and patients with high circulating MIF (>median) 12 hours after surgery had a significantly reduced risk of developing AKI (relative risk reduction, 72.7%; 95% confidence interval, 12 to 91.5%; P = 0.03). Experimental AKI was induced in wild-type and Mif-/- mice by 30 min of ischemia followed by 6 or 24 hours of reperfusion, or by rhabdomyolysis. Mif-deficient mice exhibited increased tubular cell injury, increased regulated cell death (necroptosis and ferroptosis), and enhanced oxidative stress. Therapeutic administration of recombinant MIF after ischemia-reperfusion in mice ameliorated AKI. In vitro treatment of tubular epithelial cells with recombinant MIF reduced cell death and oxidative stress as measured by glutathione and thiobarbituric acid reactive substances in the setting of hypoxia. Our data provide evidence of a renoprotective role of MIF in experimental ischemia-reperfusion injury by protecting renal tubular epithelial cells, consistent with our observation that high MIF in cardiac surgery patients is associated with a reduced incidence of AKI.


Sujet(s)
Atteinte rénale aigüe/sang , Atteinte rénale aigüe/étiologie , Procédures de chirurgie cardiaque/effets indésirables , Facteurs inhibiteurs de la migration des macrophages/sang , Facteurs inhibiteurs de la migration des macrophages/urine , Agents protecteurs/métabolisme , Atteinte rénale aigüe/épidémiologie , Atteinte rénale aigüe/urine , Animaux , Antigènes de différenciation des lymphocytes B/composition chimique , Antigènes de différenciation des lymphocytes B/métabolisme , Antioxydants/métabolisme , Mort cellulaire , Antigènes d'histocompatibilité de classe II/composition chimique , Antigènes d'histocompatibilité de classe II/métabolisme , Humains , Incidence , Inflammation/anatomopathologie , Rein/vascularisation , Rein/anatomopathologie , Peroxydation lipidique , Lipocaline-2/urine , Facteurs inhibiteurs de la migration des macrophages/déficit , Souris de lignée C57BL , Stress oxydatif , Domaines protéiques , Protéines recombinantes/administration et posologie , Protéines recombinantes/pharmacologie , Lésion d'ischémie-reperfusion/complications , Lésion d'ischémie-reperfusion/anatomopathologie , Rhabdomyolyse/anatomopathologie
12.
Int J Mol Sci ; 17(7)2016 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-27447611

RÉSUMÉ

Increasing evidence indicates that argon has organoprotective properties. So far, the underlying mechanisms remain poorly understood. Therefore, we investigated the effect of argon preconditioning in cardiomyocytes within the first and second window of preconditioning. Primary isolated cardiomyocytes from neonatal rats were subjected to 50% argon for 1 h, and subsequently exposed to a sublethal dosage of hypoxia (<1% O2) for 5 h either within the first (0-3 h) or second window (24-48 h) of preconditioning. Subsequently, the cell viability and proliferation was measured. The argon-induced effects were assessed by evaluation of mRNA and protein expression after preconditioning. Argon preconditioning did not show any cardioprotective effects in the early window of preconditioning, whereas it leads to a significant increase of cell viability 24 h after preconditioning compared to untreated cells (p = 0.015) independent of proliferation. Argon-preconditioning significantly increased the mRNA expression of heat shock protein (HSP) B1 (HSP27) (p = 0.048), superoxide dismutase 2 (SOD2) (p = 0.001), vascular endothelial growth factor (VEGF) (p < 0.001) and inducible nitric oxide synthase (iNOS) (p = 0.001). No difference was found with respect to activation of pro-survival kinases in the early and late window of preconditioning. The findings provide the first evidence of argon-induced effects on the survival of cardiomyocytes during the second window of preconditioning, which may be mediated through the induction of HSP27, SOD2, VEGF and iNOS.


Sujet(s)
Argon/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytoprotection/effets des médicaments et des substances chimiques , Cardiopathies/prévention et contrôle , Préconditionnement ischémique myocardique , Myocytes cardiaques/effets des médicaments et des substances chimiques , Animaux , Technique de Western , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines du choc thermique/génétique , Protéines du choc thermique/métabolisme , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Rats , Rat Wistar , Réaction de polymérisation en chaine en temps réel , Superoxide dismutase/génétique , Superoxide dismutase/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme
13.
J Cardiovasc Transl Res ; 9(3): 230-238, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-27055858

RÉSUMÉ

In the present observational study, we measured serum levels of the chemokine stromal cell-derived factor-1α (SDF-1α) in 100 patients undergoing cardiac surgery with cardiopulmonary bypass at seven distinct time points including preoperative values, myocardial ischemia, reperfusion, and the postoperative course. Myocardial ischemia triggered a marked increase of SDF-1α serum levels whereas cardiac reperfusion had no significant influence. Perioperative SDF-1α serum levels were influenced by patients' characteristics (e.g., age, gender, aspirin intake). In an explorative analysis, we observed an inverse association between SDF-1α serum levels and the incidence of organ dysfunction. In conclusion, time of myocardial ischemia was identified as the key stimulus for a significant upregulation of SDF-1α, indicating its role as a marker of myocardial injury. The inverse association between SDF-1α levels and organ dysfunction association encourages further studies to evaluate its organoprotective properties in cardiac surgery patients.


Sujet(s)
Chimiokine CXCL12/sang , Pontage aortocoronarien/effets indésirables , Maladie des artères coronaires/chirurgie , Valvulopathies/chirurgie , Implantation de valve prothétique cardiaque/effets indésirables , Lésion de reperfusion myocardique/sang , Sujet âgé , Marqueurs biologiques/sang , Hypoxie cellulaire , Cellules cultivées , Maladie des artères coronaires/sang , Maladie des artères coronaires/diagnostic , Femelle , Valvulopathies/sang , Valvulopathies/diagnostic , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Mâle , Adulte d'âge moyen , Lésion de reperfusion myocardique/diagnostic , Lésion de reperfusion myocardique/étiologie , Valeur prédictive des tests , Facteurs de risque , Facteurs temps , Résultat thérapeutique
14.
J Cell Mol Med ; 20(1): 104-15, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26499307

RÉSUMÉ

The chemokine CXCL16 and its receptor CXCR6 have been linked to the pathogenesis of acute and chronic cardiovascular disease. However, data on the clinical significance of CXCL16 in patients undergoing cardiac surgery with acute myocardial ischemia/reperfusion (I/R) are still lacking. Therefore, we determined CXCL16 in the serum of cardiac surgery patients and investigated its kinetics and association with the extent of organ dysfunction. 48 patients underwent conventional cardiac surgery with myocardial I/R and the use of cardiopulmonary bypass (CPB) were consecutively enrolled in the present study. We investigated the peri- and post-operative profile of CXCL16. Clinical relevant data were assessed and documented throughout the entire observation period. To identify the influence of myocardial I/R and CPB on CXCL16 release data were compared to those received from patients that underwent off-pump procedure. Pre-operative serum CXCL16 levels were comparable to those obtained from healthy volunteers (1174 ± 55.64 pg/ml versus 1225 ± 70.94). However, CXCL16 levels significantly increased during surgery (1174 ± 55.64 versus 1442 ± 75.42 pg/ml; P = 0.0057) and reached maximum levels 6 hrs after termination of surgery (1174 ± 55.64 versus 1648 ± 74.71 pg/ml; P < 0.001). We revealed a positive correlation between the intraoperative serum levels of CXCL16 and the extent of organ dysfunction (r(2) = 0.356; P = 0.031). Patients with high CXCL16 release showed an increased extent of organ dysfunction compared to patients with low CXCL16 release. Our study shows that CXCL16 is released into the circulation as a result of cardiac surgery and that high post-operative CXCL16 levels are associated with an increased severity of post-operative organ dysfunctions.


Sujet(s)
Chimiokines CXC/sang , Cardiopathies/sang , Récepteurs éboueurs/sang , Sujet âgé , Cellules cultivées , Chimiokine CXCL16 , Femelle , Cardiopathies/immunologie , Cardiopathies/chirurgie , Humains , Numération des leucocytes , Mâle , Période périopératoire , Complications postopératoires/sang , Syndrome de réponse inflammatoire généralisée/sang , Résultat thérapeutique
15.
Antioxid Redox Signal ; 23(11): 865-79, 2015 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-26234719

RÉSUMÉ

AIMS: Cardiac surgery involves myocardial ischemia/reperfusion (I/R) with potentially deleterious consequences. Macrophage migration inhibitory factor (MIF) is a stress-regulating chemokine-like cytokine that protects against I/R damage, but functional links with its homolog, d-dopachrome tautomerase (MIF-2), and the circulating soluble receptor CD74 (sCD74) are unknown. In this study, we investigate the role of MIF, MIF-2, sCD74, and MIF genotypes in patients scheduled for elective single or complex surgical procedures such as coronary artery bypass grafting or valve replacement. RESULTS: MIF and MIF-2 levels significantly increased intraoperatively, whereas measured sCD74 decreased correspondingly. Circulating sCD74/MIF complexes were detectable in 50% of patients and enhanced MIF antioxidant activity. Intraoperative MIF levels were independently associated with a reduced risk for the development of atrial fibrillation (AF) (odds ratio 0.99 [0.98-1.00]; p=0.007). Circulating levels of MIF-2, but not MIF, were associated with an increased frequency of organ dysfunction and predicted the occurrence of AF (area under the curve [AUC]=0.663; p=0.041) and pneumonia (AUC=0.708; p=0.040). Patients with a high-expression MIF genotype exhibited a reduced incidence of organ dysfunction compared with patients with low-expression MIF genotypes (3 vs. 25; p=0.042). INNOVATION: The current study comprehensively highlights the kinetics and clinical relevance of MIF family proteins and the MIF genotype in cardiac surgery patients. CONCLUSION: Our findings suggest that increased MIF levels during cardiac surgery feature organ-protective properties during myocardial I/R, while the soluble MIF receptor, sCD74, may enhance MIF antioxidant activity. In contrast, high MIF-2 levels are predictive of the development of organ dysfunction. Importantly, we provide first evidence for a gene-phenotype relationship between variant MIF alleles and clinical outcome in cardiac surgery patients.


Sujet(s)
Intramolecular oxidoreductases/sang , Facteurs inhibiteurs de la migration des macrophages/sang , Lésion de reperfusion myocardique/sang , Sujet âgé , Animaux , Antigènes de différenciation des lymphocytes B/sang , Marqueurs biologiques/sang , Mouvement cellulaire , Cellules cultivées , Pontage aortocoronarien , Femelle , Antigènes d'histocompatibilité de classe II/sang , Humains , Intramolecular oxidoreductases/génétique , Facteurs inhibiteurs de la migration des macrophages/génétique , Mâle , Adulte d'âge moyen , Granulocytes neutrophiles/physiologie , Oxydoréduction , Polymorphisme de nucléotide simple , Période postopératoire , Facteurs de protection , Rats , Résultat thérapeutique
16.
PLoS One ; 9(3): e92827, 2014.
Article de Anglais | MEDLINE | ID: mdl-24667295

RÉSUMÉ

INTRODUCTION: Anesthetic-induced preconditioning (AIP) is known to elicit cardioprotective effects that are mediated at least in part by activation of the kinases AMPK and PKCε as well as by inhibition of JNK. Recent data demonstrated that the pleiotropic cytokine macrophage migration inhibitory factor (MIF) provides cardioprotection through activation and/or inhibition of kinases that are also known to mediate effects of AIP. Therefore, we hypothesized that MIF could play a key role in the AIP response. METHODS: Cardiomyocytes were isolated from rats and subjected to isoflurane preconditioning (4 h; 1.5 vol. %). Subsequently, MIF secretion and alterations in the activation levels of protective kinases were compared to a control group that was exposed to ambient air conditions. MIF secretion was quantified by ELISA and AIP-induced activation of protein kinases was assessed by Western blotting of cardiomyocyte lysates after isoflurane treatment. RESULTS: In cardiomyocytes, preconditioning with isoflurane resulted in a significantly elevated secretion of MIF that followed a biphasic behavior (30 min vs. baseline: p = 0.020; 24 h vs. baseline p = 0.000). Moreover, quantitative polymerase chain reaction demonstrated a significant increase in MIF mRNA expression 8 h after AIP. Of note, activation of AMPK and PKCε coincided with the observed peaks in MIF secretion and differed significantly from baseline. CONCLUSIONS: These results suggest that the pleiotropic mediator MIF is involved in anesthetic-induced preconditioning of cardiomyocytes through stimulation of the protective kinases AMPK and PKCε.


Sujet(s)
Anesthésiques par inhalation/pharmacologie , Cardiotoniques/pharmacologie , Intramolecular oxidoreductases/métabolisme , Isoflurane/pharmacologie , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Protéines du muscle/métabolisme , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Animaux , Cellules cultivées , Activation enzymatique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Myocarde/cytologie , Myocytes cardiaques/cytologie , Protein kinases/métabolisme , Rats , Rat Wistar
17.
Mini Rev Med Chem ; 14(14): 1116-24, 2014.
Article de Anglais | MEDLINE | ID: mdl-25643612

RÉSUMÉ

Emerging evidence suggests a pivotal role of macrophage migration inhibitory factor (MIF) in the systemic inflammatory immune response. MIF is located in various cell types and rapidly released after different stimuli like inflammation, surgical stress or ischemia and reperfusion. MIF is a known key player in the inflammatory response and contributes to several biological functions including the control of cell cycle (through activation of ERK1/2), sensing of pathogen stimuli (upregulation of TLR4 expression), recruitment of various immune cells (neutrophils, monocytes) and prevention of p53-mediated apoptosis of macrophages. While MIF`s pro-inflammatory effects are crucial for an effective host defense, elevated MIF levels were repeatedly shown to be associated with the development of organ dysfunction and deleterious sequelae. Even more puzzling, increasing evidence indicates a protective role of this pleiotropic cytokine during ischemia and reperfusion injury in the myocardium. This review focuses on new insights regarding the biological significance of MIF release in the context of critical illness and ischemia/ reperfusion.


Sujet(s)
Maladie grave , Facteurs inhibiteurs de la migration des macrophages/physiologie , Récepteurs immunologiques/physiologie , Lésion d'ischémie-reperfusion/métabolisme , Humains , Médiateurs de l'inflammation/physiologie , Lésion d'ischémie-reperfusion/diagnostic , Lésion d'ischémie-reperfusion/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...