Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 170: 115993, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38091635

ABSTRACT

Cardiomyopathy is a common complication and significantly increases the risk of death in septic patients. Our previous study demonstrated that post-treatment with dexmedetomidine (DEX) aggravates septic cardiomyopathy. However, the mechanisms for the side effect of DEX post-treatment on septic cardiomyopathy are not well-defined. Here we employed a cecal ligation and puncture (CLP) model and α2A-adrenoceptor deficient (Adra2a-/-) mice to observe the effects of DEX post-treatment on myocardial metabolic disturbances in sepsis. CLP mice displayed significant cardiac dysfunction, altered mitochondrial dynamics, reduced cardiac lipid and glucose uptake, impaired fatty acid and glucose oxidation, enhanced glycolysis and decreased ATP production in the myocardium, almost all of which were dramatically enhanced by DEX post-treatment in septic mice. In Adra2a-/- mice, DEX post-treatment did not affect cardiac dysfunction and metabolic disruptions in CLP-induced sepsis. Additionally, Adra2a-/- mice exhibited impaired cardiac function, damaged myocardial mitochondrial structures, and disturbed fatty acid metabolism and glucose oxidation. In sum, DEX post-treatment exacerbates metabolic disturbances in septic cardiomyopathy in a α2A-adrenoceptor dependent manner.


Subject(s)
Cardiomyopathies , Dexmedetomidine , Heart Diseases , Sepsis , Humans , Mice , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Cardiomyopathies/drug therapy , Heart Diseases/drug therapy , Sepsis/drug therapy , Glucose/therapeutic use , Fatty Acids
2.
Int Immunopharmacol ; 116: 109724, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36696856

ABSTRACT

BACKGROUND: Dexmedetomidine (DEX) administered before or at 30 min after sepsis induction was reported to alleviate septic cardiomyopathy in experimental models. However, sepsis is a life-threatening organ dysfunction due to infection-induced dysregulated host response, whether DEX treatment in the presence of organ dysfunction affects septic cardiomyopathy is unknown. This study investigated the effect of DEX posttreatment on septic cardiomyopathy. METHODS: Male wild-type and α2A-adrenergic receptor (AR) knockout mice were exposed to lipopolysaccharide (LPS) or cecal ligation puncture (CLP), and cultured cardiac endothelial cells were used. Mouse survival, myocardial function, inflammatory response and related signaling pathways were determined. RESULTS: DEX treatment at 6, 9 h after LPS challenge significantly reduced survival rate of LPS-challenged mice, especially at 9 h. DEX administered at 9 h after LPS injection or CLP significantly reduced survival in LPS or CLP-induced sepsis in wild-type mice, but not in α2A-AR knockout mice. LPS treatment for 20 h decreased the left ventricle + dp/dt, increased myocardial interleukin (IL)-1ß and IL-6 concentrations as well as cardiac endothelial tumor necrosis factor (TNF)-α, vascular cell adhesion molecule-1 (VCAM-1) and ICAM-1 expression, which were enhanced by DEX treated at 9 h after LPS injection in wild-type mice, but not in α2A-AR knockout mice. Furthermore, DEX posttreatment increased p38 phosphorylation, c-Fos nuclear translocation and VCAM-1 expression in LPS-treated cardiac endothelial cells, which were eliminated by α2A-AR knockout or PKC inhibitor. CONCLUSIONS: DEX posttreatment aggravates LPS-induced cardiac inflammation and myocardial dysfunction, at least in part, via activating cardiac endothelial α2A-AR-mediated PKC signal pathway.


Subject(s)
Cardiomyopathies , Dexmedetomidine , Sepsis , Mice , Male , Animals , Lipopolysaccharides/pharmacology , Dexmedetomidine/therapeutic use , Dexmedetomidine/pharmacology , Endothelial Cells/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Multiple Organ Failure , Tumor Necrosis Factor-alpha/metabolism , Sepsis/drug therapy , Mice, Knockout
3.
Commun Biol ; 5(1): 96, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35079095

ABSTRACT

Intrinsic cardiac adrenergic (ICA) cells regulate both developing and adult cardiac physiological and pathological processes. However, the role of ICA cells in septic cardiomyopathy is unknown. Here we show that norepinephrine (NE) secretion from ICA cells is increased through activation of Toll-like receptor 4 (TLR4) to aggravate myocardial TNF-α production and dysfunction by lipopolysaccharide (LPS). In ICA cells, LPS activated TLR4-MyD88/TRIF-AP-1 signaling that promoted NE biosynthesis through expression of tyrosine hydroxylase, but did not trigger TNF-α production due to impairment of p65 translocation. In a co-culture consisting of LPS-treated ICA cells and cardiomyocytes, the upregulation and secretion of NE from ICA cells activated cardiomyocyte ß1-adrenergic receptor driving Ca2+/calmodulin-dependent protein kinase II (CaMKII) to crosstalk with NF-κB and mitogen-activated protein kinase pathways. Importantly, blockade of ICA cell-derived NE prevented LPS-induced myocardial dysfunction. Our findings suggest that ICA cells may be a potential therapeutic target for septic cardiomyopathy.


Subject(s)
Cardiomyopathies/chemically induced , Cardiovascular Agents/pharmacology , Lipopolysaccharides/toxicity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Norepinephrine/metabolism , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/metabolism , Gene Deletion , Gene Expression Regulation/drug effects , Male , Mice , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4
4.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884552

ABSTRACT

Dexmedetomidine (DEX), a selective α2 adrenergic receptor (AR) agonist, is commonly used as a sedative drug during critical illness. In the present study, we explored a novel accelerative effect of DEX on cardiac fibroblast (CF) differentiation mediated by LPS and clarified its potential mechanism. LPS apparently increased the expression of α-SMA and collagen I/III and the phosphorylation of p38 and Smad-3 in the CFs of mice. These effects were significantly enhanced by DEX through increasing α2A-AR expression in CFs after LPS stimulation. The CFs from α2A-AR knockout mice were markedly less sensitive to DEX treatment than those of wild-type mice. Inhibition of protein kinase C (PKC) abolished the enhanced effects of DEX on LPS-induced differentiation of CFs. We also found that the α-SMA level in the second-passage CFs was much higher than that in the nonpassage and first-passage CFs. However, after LPS stimulation, the TNF-α released from the nonpassage CFs was much higher than that in the first- and second-passage CFs. DEX had no effect on LPS-induced release of TNF-α and IL-6 from CFs. Further investigation indicated that DEX promoted cardiac fibrosis and collagen I/III synthesis in mice exposed to LPS for four weeks. Our results demonstrated that DEX effectively accelerated LPS-induced differentiation of CFs to myofibroblasts through the PKC-p38-Smad2/3 signaling pathway by activating α2A-AR.


Subject(s)
Cell Differentiation , Collagen Type III/metabolism , Collagen Type I/metabolism , Dexmedetomidine/pharmacology , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Myofibroblasts/cytology , Receptors, Adrenergic, alpha-2/chemistry , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Shock ; 56(4): 582-592, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34524268

ABSTRACT

ABSTRACT: Dobutamine (DOB) is recommended as an inotrope for septic patients with low cardiac output, but its long-term impact on sepsis-induced cardiomyopathy remains unclear. This study investigated the long-term effect of DOB on septic myocardial dysfunction and injury. Rats were exposed to cecal ligation and puncture (CLP), the intrinsic myocardial function, other organ functions, hemodynamics, inflammatory response, serum myocardial injury biomarkers, myocardial apoptosis, and vascular permeability were determined. At 6 h after CLP, the left ventricular ±dP/dt were significantly depressed, cardiac tumor necrosis factor-α and vascular cell adhesion molecule-1 expression were increased, but not serum cardiac troponin I (cTnI), N-terminal pro-brain natriuretic peptide (NT-proBNP), heart-type fatty acid-binding protein (H-FABP), creatinine, and urea nitrogen concentrations in CLP group compared with controls. At 9 h after CLP, hepatic dysfunction was present in CLP rats compared with controls. At 6 h after CLP, DOB treatment did not affect hemodynamics, the left ventricular ±dP/dt, cytokine levels in serum and myocardium, as well as cardiomyocyte apoptosis and cardiac vascular hyperpermeability at 20 h after CLP. However, DOB (10.0 µg/kg) increased serum IL-10 level and improved survival in septic rats. These results indicate that the intrinsic myocardial depression occurs earlier than hepatic and renal dysfunction in sepsis and serum cTnI, NT-proBNP, and H-FABP are not suitable as early biomarkers for sepsis-induced myocardial dysfunction. Although DOB treatment (10.0 µg/kg) in the presence of myocardial dysfunction improves survival in septic rats, it neither improves myocardial function and hemodynamics nor attenuates myocardial injury at the later stage of sepsis.


Subject(s)
Cardiomyopathies/drug therapy , Cardiotonic Agents/therapeutic use , Dobutamine/therapeutic use , Heart Injuries/drug therapy , Sepsis/complications , Animals , Cardiomyopathies/etiology , Cytokines/blood , Disease Models, Animal , Heart Injuries/etiology , Male , Rats , Rats, Sprague-Dawley , Time Factors
6.
J Cardiovasc Pharmacol ; 73(3): 186-194, 2019 03.
Article in English | MEDLINE | ID: mdl-30839512

ABSTRACT

OBJECTIVE: To investigate whether phenylephrine (PE) inhibits sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury through the PI3K/Akt signaling pathway. METHODS: A rat model of sepsis was established by cecal ligation and puncture. PE and/or wortmannin (a PI3K inhibitor) were administered to investigate the role of PI3K/Akt signaling in mediating the effects of PE on inhibiting sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury. Hematoxylin-eosin staining, echocardiography, and Langendorff system were used to examine the myocardial injury and function. The concentrations of TNF-α and IL-6 were analyzed by enzyme-linked immunosorbent assay. Intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), myeloperoxidase, mitochondria-related fusion/fission proteins, and PI3K/Akt signaling pathway-associated proteins were analyzed by Western blotting. RESULTS: PE improved the cardiac function and survival in septic rats. PE decreased TNF-α, IL-6, ICAM-1, VCAM-1, and myeloperoxidase contents in the myocardium of septic rats. Meanwhile, PE increased the fusion-related proteins and decreased the fission-related proteins in the myocardial mitochondria of septic rats. On the other hand, PE activated the PI3K/Akt signaling pathway in the cecal ligation and puncture-treated rats, and all the protective effects of PE were abolished by wortmannin. CONCLUSIONS: PE attenuated sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury through the PI3K/Akt signaling pathway.


Subject(s)
Mitochondria, Heart/drug effects , Mitochondrial Dynamics/drug effects , Myocarditis/prevention & control , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/drug therapy , Animals , Disease Models, Animal , Inflammation Mediators/metabolism , Isolated Heart Preparation , Male , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Mitochondrial Proteins/metabolism , Myocarditis/enzymology , Myocarditis/etiology , Myocarditis/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Peroxidase/metabolism , Rats, Sprague-Dawley , Sepsis/complications , Signal Transduction , Stroke Volume/drug effects , Ventricular Function, Left
7.
Shock ; 52(1): 122-133, 2019 07.
Article in English | MEDLINE | ID: mdl-29889817

ABSTRACT

It was demonstrated that α1 adrenergic receptor (α1-AR) activation by phenylephrine (PE) attenuated cardiac dysfunction in lipopolysaccharide (LPS)-challenged mice. However, it is unclear whether PE suppresses sepsis-induced cardiomyocyte apoptosis. Here, we investigated the effects of PE on cardiomyocyte apoptosis in LPS-treated adult rat ventricular myocytes (ARVMs) and septic rats induced by cecal ligation and puncture. Cardiomyocyte apoptosis and caspase activity were detected by TUNEL and spectrophotometrical assay, respectively. Bax, Bcl-2 and cytochrome c (Cyt c) levels as well as IκBα, ERK1/2, p38 MAPK, JNK and cardiac troponin I (cTnI) phosphorylation were analyzed by Western blotting, and TNF-α concentration was analyzed by ELISA. PE inhibited LPS-induced caspase-3 activation in ARVMs, which was reversed by prazosin (a membrane permeable α1-AR antagonist), but not by CGP12177A (a membrane impermeable α1-AR antagonist). PE upregulated phosphorylated ERK1/2 and Bcl-2 contents, decreased TNF-α and Bax levels, Cyt c release, caspase-8/-9 activities as well as IκBα, p38MAPK and JNK phosphorylation in LPS-treated ARVMs, all of which were abolished by prazosin. Treatment with U0126 (a specific ERK1/2 inhibitor) reversed the effects of PE on IκBα, p38MAPK and JNK phosphorylation as well as caspase-3/-8/-9 activation in LPS-treated ARVMs. In septic rats, PE not only inhibited myocardial apoptosis as well as IκBα, p38MAPK, and JNK phosphorylation, but also upregulated myocardial phosphorylated ERK1/2. Furthermore, PE inhibited myocardial cTnI phosphorylation and improved cardiac function in septic rats. Taken together, our data suggest that α1-AR activation by PE inhibits sepsis-induced cardiomyocyte apoptosis and cardiac dysfunction via activating ERK1/2 signal pathway.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/therapeutic use , Lipopolysaccharides/toxicity , Phenylephrine/therapeutic use , Sepsis/drug therapy , Sepsis/physiopathology , Animals , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cytochromes c/metabolism , Enzyme-Linked Immunosorbent Assay , In Situ Nick-End Labeling , Male , Myocytes, Cardiac/drug effects , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Troponin T/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Int J Cardiol ; 270: 293-301, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-29908831

ABSTRACT

BACKGROUND: Neonatal rat ventricular myocytes (NRVMs) have proven to be an ideal research model for cardiac disease. However, the current methods to purify NRVMs have a limitation to obtain high purity. The purpose of this study was to develop a NRVM purification method by using superparamagnetic iron oxide particles (SIOP). METHODS: NRVMs were purified by using SIOP (SIOP group). The differential attachment with or without bromodeoxyuridine (BrdU) treatment served as control and BrdU groups, respectively. The Percoll gradient (Percoll) and magnetic-activated cell sorting (MACS) methods were performed to compare the purity and viability of NRVMs with SIOP method. RESULTS: The SIOP group enriched NRVMs up to 93.9 ±â€¯2.0% purity determined by flow cytometry (FCM) and 95.6 ±â€¯1.3% by immunofluorescence count (IF). In contrast, the control group gave purities of 71.9 ±â€¯2.9% (by FCM) and 66.8 ±â€¯8.9% (by IF), and the BrdU group obtained 82.0 ±â€¯1.3% (by FCM) and 83.1 ±â€¯2.4% (by IF). The purity of SIOP-isolated NRVMs was not different from that of Percoll and MACS groups. However, the cardiomyocytes separated by these methods, except SIOP protocol, were mixed with intrinsic cardiac adrenergic cells. NRVMs purified by SIOP shaped the similar three-dimensional morphology, with no difference in cell yield, viability and cytosolic Ca2+ homeostasis at 24 h after isolation compared with NRVMs in other groups. Furthermore, SIOP-purified NRVMs retained the responses to phenylephrine and lipopolysaccharide challenge. CONCLUSION: We first reported an efficient and novel method to purify NRVMs using SIOP, which may help accelerate innovative research in the field of cardiomyocyte biology.


Subject(s)
Cell Separation/methods , Ferric Compounds/administration & dosage , Heart Ventricles/cytology , Heart Ventricles/drug effects , Magnetite Nanoparticles/administration & dosage , Myocytes, Cardiac/drug effects , Animals , Animals, Newborn , Cells, Cultured , Myocytes, Cardiac/physiology , Rats , Rats, Sprague-Dawley
9.
Sci Rep ; 8(1): 5478, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29615637

ABSTRACT

Cardiomyopathy is a common complication associated with increased mortality in sepsis, but lacks specific therapy. Here, using genetic and pharmacological approaches, we explored the therapeutic effect of α2A-adrenergic receptor (AR) blockade on septic cardiomyopathy. CLP-induced septic rats were treated with BRL44408 (α2A-AR antagonist), prazosin (α1-AR antagonist) and/or reserpine. CLP-induced cardiomyopathy, indicated by reduced dP/dt and increased cardiac troponin I phosphorylation, was attenuated by BRL44408, this was associated with reduced cardiac TNF-α and endothelial VCAM-1 expression, cardiomyocyte apoptosis and related signal molecule phosphorylation. BRL44408 increased cardiac norepinephrine (NE) concentration in CLP rats. Pretreatment with reserpine that exhausts cardiac NE without affecting the circulating NE concentration or with prazosin partially abolished the cardioprotection of BRL44408 and reversed its inhibitory effects on myocardial TNF-α, apoptosis and related signal molecule phosphorylation, but not on VCAM-1 expression in septic rats. These effects of BRL44408 were confirmed by α2A-AR gene deletion in septic mice. Furthermore, α2-AR agonist not only enhanced LPS-induced TNF-α and VCAM-1 expression in cardiac endothelial cells that express α2A-AR, but also enhanced LPS-induced cardiac dysfunction in isolated rat hearts. Our data indicate that α2A-AR blockade attenuates septic cardiomyopathy by promoting cardiac NE release that activates myocardial α1-AR and suppressing cardiac endothelial activation.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Cardiomyopathies/drug therapy , Endothelial Cells/drug effects , Myocardium/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Sepsis/complications , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Apoptosis/drug effects , Cardiomyopathies/complications , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Deletion , Gene Expression Regulation/drug effects , Heart/drug effects , Heart/physiopathology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Myocardium/pathology , NF-KappaB Inhibitor alpha/metabolism , Neutrophil Infiltration/drug effects , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/deficiency , Receptors, Adrenergic, alpha-2/genetics , Survival Analysis , Tumor Necrosis Factor-alpha/biosynthesis , Vascular Cell Adhesion Molecule-1/metabolism , Ventricular Function, Left/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Int Immunopharmacol ; 35: 217-225, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27082997

ABSTRACT

Infiltration of activated neutrophils into the vital organs contributes to the multiple organ dysfunctions in sepsis. In the present study, we investigated the effects of berberine in combination with yohimbine (BY) on neutrophil tissue infiltration and multiple organ damage during sepsis, and further elucidated the involved mechanisms. Sepsis was induced in mice by cecal ligation and puncture (CLP). BY or CCR2 antagonist was administered 2h after CLP, and anti-IL-10 antibody (IL-10 Ab) or control IgG was injected intraperitoneally just before BY treatment. We found that IL-10 production was enhanced by BY therapy in septic mice. BY significantly attenuated neutrophil tissue infiltration and multiple organ injury in CLP-challenged mice, all of which were completely reversed by IL-10 Ab pretreatment. The levels of KC, MCP-1, MIP-1α and MIP-2 in the lung, liver and kidney were markedly increased 6h after CLP. BY reduced the tissue concentrations of these chemokines in septic mice, but IL-10 Ab pretreatment did not completely eliminate these inhibitory effects of BY. Particularly, dramatically increased CCR2 expression in circulating neutrophils of septic mice was reduced by BY and this effect was completely abolished by IL-10 Ab pretreatment. Furthermore, CCR2 antagonist also inhibited lung and renal injury and neutrophil infiltration in septic mice. Taken together, our data strongly suggest that BY therapy attenuates neutrophil tissue infiltration and multiple organ injury in septic mice, at least in part, via IL-10-mediated inhibition of CCR2 expression in circulating neutrophils.


Subject(s)
Berberine/therapeutic use , Interleukin-10/metabolism , Multiple Organ Failure/drug therapy , Neutrophils/drug effects , Receptors, CCR2/metabolism , Sepsis/drug therapy , Yohimbine/therapeutic use , Animals , Antibodies, Blocking/administration & dosage , Cecum/surgery , Cell Movement/drug effects , Cells, Cultured , Disease Models, Animal , Drug Therapy, Combination , Humans , Interleukin-10/immunology , Male , Mice , Mice, Inbred Strains , Neutrophils/physiology , Receptors, CCR2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...