Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 11004, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358823

ABSTRACT

Cardiac fibrosis is an underlying cause of diastolic dysfunction, contributing to heart failure. Substance P (SP) activation of the neurokinin-1 receptor (NK-1R) contributes to cardiac fibrosis in hypertension. However, based on in vitro experiments, this does not appear to be via direct activation of cardiac fibroblasts. While numerous cells could mediate the fibrotic effects of SP, herein, we investigate mast cells (MC) as a mechanism mediating the fibrotic actions of SP, since MCs are known to play a role in cardiac fibrosis and respond to SP. Spontaneously hypertensive rats (SHR) were treated with the NK-1R antagonist L732138 (5 mg/kg/d) from 8 to 12 weeks of age. L732138 prevented increased MC maturation of resident immature MCs. NK-1R blockade also prevented increased cardiac MC maturation in angiotensin II-infused mice. MC-deficient mice were used to test the importance of MC NK-1Rs to MC activation. MC-deficient mice administered angiotensin II did not develop fibrosis; MC-deficient mice reconstituted with MCs did develop fibrosis. MC-deficient mice reconstituted with MCs lacking the NK-1R also developed fibrosis, indicating that NK-1Rs are not required for MC activation in this setting. In conclusion, the NK-1R causes MC maturation, however, other stimuli are required to activate MCs to cause fibrosis.


Subject(s)
Mast Cells/pathology , Myocardium/pathology , Receptors, Neurokinin-1/metabolism , 3T3 Cells , Animals , Apoptosis , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Hypertension/metabolism , Hypertension/pathology , Male , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Myocardium/cytology , Myocardium/metabolism , Rats, Inbred SHR , Rats, Inbred WKY
2.
Neuropeptides ; 74: 52-59, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30660328

ABSTRACT

The neuropeptide substance P can induce degranulation of cardiac mast cells at high concentrations. Herein, we seek to further understand substance P activation of cardiac mast cells in the context of other neuropeptides as well as modulation by non-neuropeptides. This is important given the increasingly recognized role of both cardiac mast cells and substance P in adverse cardiac remodeling. To address this, we isolated cardiac mast cells and compared their response to substance P as well as other members from the tachykinin family of peptides, including neurokinin A and hemokinin-1. We also tested the ability of other factors to manipulate the cardiac mast cell response to substance P. We found that while neurokinin A did not induce cardiac mast cell degranulation, both substance P and hemokinin-1 induced a concentration-dependent release of histamine; the maximal response to hemokinin-1 was greater than to substance P. Neurokinin-1 receptor blockade prevented substance P-induced histamine release, while only partially attenuating hemokinin-1-induced histamine release. The antioxidant N-acetylcysteine attenuated histamine release in response to hemokinin-1 and had no effect on substance P-induced histamine release. Selective PPAR-γ agonists attenuated histamine release in response to substance P. These data indicate that substance P activates cardiac mast cells via the neurokinin-1 receptor, and that the activation response is different to other tachykinins. That the response to substance P is receptor mediated and can be modulated by activation of other receptors (PPAR-γ), argues that substance P activation of cardiac mast cells has potential biological significance.


Subject(s)
Mast Cells/metabolism , Myocardium/metabolism , Substance P/metabolism , Animals , Cell Degranulation , Histamine/metabolism , Histamine Release , Male , Mast Cells/cytology , Myocardium/cytology , PPAR gamma/metabolism , Rats, Sprague-Dawley , Receptors, Neurokinin-1/metabolism , Substance P/administration & dosage , Ventricular Remodeling
3.
IJC Metab Endocr ; 10: 16-23, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26722642

ABSTRACT

BACKGROUND: Cardiac mast cell (MC) proteases, chymase and tryptase, increase proliferation and collagen synthesis in cultured cardiac fibroblasts. However, the question as to why preventing individually the actions of either protease prevents fibrosis when both are released upon MC activation remains unanswered. Since tryptase has the ability to activate MCs in noncardiac tissues via the protease-activated receptor-2 (PAR-2), there is the possibility that its, in vivo, fibrotic role is due to its ability to induce MC degranulation thereby amplifying the release of chymase. METHODS: This study sought to delineate the interactions between tryptase and chymase in myocardial remodeling secondary to transverse aortic constriction (TAC) for 5 wks in male Sprague Dawley rats untreated or treated with either the tryptase inhibitor, nafamostat mesilate or MC membrane stabilizing drug, nedocromil (n=6/group). In addition, ventricular slices from 6 rat hearts were incubated with tryptase, tryptase plus nafamostat mesilate or chymostatin for 24 h. RESULTS AND CONCLUSION: The results indicate the presence of PAR-2 on MCs and that tryptase inhibition and nedocromil prevented TAC-induced fibrosis and increases in MC density, activation, and chymase release. Tryptase also significantly increased chymase concentration in ventricular tissue culture media, which was prevented by the tryptase inhibitor. Hydroxyproline concentration in culture media was significantly increased with tryptase incubation as compared to the control group and the tryptase group incubated with nafamostat mesilate or chymostatin. We conclude that tryptase contributes to TAC-induced cardiac fibrosis primarily via activation of MCs and the amplified release of chymase.

4.
Hypertension ; 67(1): 107-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26573705

ABSTRACT

Nuclear factor erythroid-2-related factor 2 (Nrf2) appears to exert either a protective or detrimental effect on the heart; however, the underlying mechanism remains poorly understood. Herein, we uncovered a novel mechanism for turning off the Nrf2-mediated cardioprotection and switching on Nrf2-mediated cardiac dysfunction. In a murine model of pressure overload-induced cardiac remodeling and dysfunction via transverse aortic arch constriction, knockout of Nrf2 enhanced myocardial necrosis and death rate during an initial stage of cardiac adaptation when myocardial autophagy function is intact. However, knockout of Nrf2 turned out to be cardioprotective throughout the later stage of cardiac maladaptive remodeling when myocardial autophagy function became insufficient. Transverse aortic arch constriction -induced activation of Nrf2 was dramatically enhanced in the heart with impaired autophagy, which is induced by cardiomyocyte-specific knockout of autophagy-related gene (Atg)5. Notably, Nrf2 activation coincided with the upregulation of angiotensinogen (Agt) only in the autophagy-impaired heart after transverse aortic arch constriction. Agt5 and Nrf2 gene loss-of-function approaches in combination with Jak2 and Fyn kinase inhibitors revealed that suppression of autophagy inactivated Jak2 and Fyn and nuclear translocation of Fyn, while enhancing nuclear translocation of Nrf2 and Nrf2-driven Agt expression in cardiomyocytes. Taken together, these results indicate that the pathophysiological consequences of Nrf2 activation are closely linked with the functional integrity of myocardial autophagy during cardiac remodeling. When autophagy is intact, Nrf2 is required for cardiac adaptive responses; however, autophagy impairment most likely turns off Fyn-operated Nrf2 nuclear export thus activating Nrf2-driven Agt transcription, which exacerbates cardiac maladaptation leading to dysfunction.


Subject(s)
Myocardium/metabolism , NF-E2-Related Factor 2/metabolism , Up-Regulation , Ventricular Pressure/physiology , Ventricular Remodeling/physiology , Animals , Autophagy , Disease Models, Animal , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/metabolism , Oxidative Stress , Signal Transduction
5.
J Mol Cell Cardiol ; 89(Pt B): 268-79, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26497614

ABSTRACT

Chronic activation of the myocardial renin angiotensin system (RAS) elevates the local level of angiotensin II (Ang II) thereby inducing pathological cardiac hypertrophy, which contributes to heart failure. However, the precise underlying mechanisms have not been fully delineated. Herein we report a novel paracrine mechanism between cardiac fibroblasts (CF)s and cardiomyocytes whereby Ang II induces pathological cardiac hypertrophy. In cultured CFs, Ang II treatment enhanced exosome release via the activation of Ang II receptor types 1 (AT1R) and 2 (AT2R), whereas lipopolysaccharide, insulin, endothelin (ET)-1, transforming growth factor beta (TGFß)1 or hydrogen peroxide did not. The CF-derived exosomes upregulated the expression of renin, angiotensinogen, AT1R, and AT2R, downregulated angiotensin-converting enzyme 2, and enhanced Ang II production in cultured cardiomyocytes. In addition, the CF exosome-induced cardiomyocyte hypertrophy was blocked by both AT1R and AT2R antagonists. Exosome inhibitors, GW4869 and dimethyl amiloride (DMA), inhibited CF-induced cardiomyocyte hypertrophy with little effect on Ang II-induced cardiomyocyte hypertrophy. Mechanistically, CF exosomes upregulated RAS in cardiomyocytes via the activation of mitogen-activated protein kinases (MAPKs) and Akt. Finally, Ang II-induced exosome release from cardiac fibroblasts and pathological cardiac hypertrophy were dramatically inhibited by GW4869 and DMA in mice. These findings demonstrate that Ang II stimulates CFs to release exosomes, which in turn increase Ang II production and its receptor expression in cardiomyocytes, thereby intensifying Ang II-induced pathological cardiac hypertrophy. Accordingly, specific targeting of Ang II-induced exosome release from CFs may serve as a novel therapeutic approach to treat cardiac pathological hypertrophy and heart failure.


Subject(s)
Exosomes/metabolism , Fibroblasts/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Renin-Angiotensin System , Amiloride/pharmacology , Angiotensin II/pharmacology , Aniline Compounds/pharmacology , Animals , Animals, Newborn , Benzylidene Compounds/pharmacology , Cardiomegaly/enzymology , Cardiomegaly/pathology , Exosomes/ultrastructure , Fibroblasts/drug effects , Fibroblasts/enzymology , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Protein Array Analysis , Protein Kinase Inhibitors/pharmacology , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
Article in English | MEDLINE | ID: mdl-26089946

ABSTRACT

Atherosclerosis is a leading cause of disability and death worldwide. Research into the disease has led to many compelling hypotheses regarding the pathophysiology of atherosclerotic lesion formation and the resulting complications such as myocardial infarction and stroke. Herbal medicine has been widely used in China as well as other Asian countries for the treatment of cardiovascular diseases for hundreds of years; however, the mechanisms of action of Chinese herbal medicine in the prevention and treatment of atherosclerosis have not been well studied. In this review, we briefly describe the mechanisms of atherogenesis and then summarize the research that has been performed in recent years regarding the effectiveness and mechanisms of antiatherogenic Chinese herbal compounds in an attempt to build a bridge between traditional Chinese medicine and cellular and molecular cardiovascular medicine.

7.
Am J Physiol Heart Circ Physiol ; 309(4): H676-84, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26071541

ABSTRACT

Accumulating evidence indicates that substance P is cardioprotective following ischemia-reperfusion primarily due to its potent coronary vasodilator actions. However, an anti-apoptotic effect of substance P has been observed in tenocytes following ischemia, which involved activation of the AKT pathway. This suggests the possibility that substance P also provides cardioprotection via direct actions to activate AKT in myocardial cells. The purpose of this study was to test the hypothesis that substance P attenuates ischemia-related cell death via direct effects on myocardial cells by activating cell survival pathways. Seven-week-old male Sprague-Dawley rats, anesthetized with intraperitoneal pentobarbital sodium (100 mg/kg), were used. The ability of substance P to prevent cellular damage was assessed following ischemia-reperfusion in an isolated heart preparation and in short-term hypoxia without reperfusion using a left ventricular tissue slice culture preparation. In addition, the NK-1 receptor and AKT involvement was assessed using the NK-1 receptor antagonist L732138 and the AKT inhibitor LY294002. The results indicate that substance P reduced the ischemia-related release of lactate dehydrogenase in both preparations and the degree of apoptosis and necrosis in the hypoxic left ventricular slices, indicating its ability to attenuate cell damage; and induced AKT phosphorylation, with both the AKT inhibitor and NK-1 receptor antagonist preventing the increased phosphorylation of AKT and the ability of substance P to attenuate hypoxic cellular damage. It is concluded that substance P reduces ischemia/hypoxia-induced myocardial cell death by acting directly on cardiac cells to initiate cell survival pathways via the NK-1 receptor and AKT.


Subject(s)
Cardiotonic Agents/pharmacology , Myocardial Reperfusion Injury/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Substance P/pharmacology , Animals , Cardiotonic Agents/therapeutic use , Cell Hypoxia , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/metabolism , Signal Transduction , Substance P/therapeutic use
8.
J Mol Cell Cardiol ; 84: 143-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25935309

ABSTRACT

Ubiquitin proteasome system (UPS) consists of ubiquitin, ubiquitin-activating enzymes (E1s), ubiquitin-conjugating enzymes (E2s), ubiquitin ligases (E3s), proteasomes, and deubiquitinating enzymes (DUBs). Ubiquitin, E1s, several E2s, E3s, and proteasomes play an important role in the regulation of cardiac homeostasis and dysfunction; however, less is known about the role of DUBs in the heart. Here, we uncovered a crucial role of cyclindromatosis (CYLD), a DUB, in mediating cardiac maladaptive remodeling and dysfunction. CYLD expression was dramatically upregulated in the cardiomyocytes of hypertrophic and failing human and murine hearts. Knockout of CYLD improved survival rate and alleviated cardiac hypertrophy, fibrosis, apoptosis, oxidative stress, and dysfunction in mice that were subjected to sustained pressure overload induced by transverse aortic constriction. Deep sequencing and gene array analyses revealed that the most dramatically changed genes are those involving in the free radical scavenging pathway and cardiovascular disease, including fos, jun, myc, and nuclear factor erythroid-2 related factor 2 (Nrf2) in the heart. Moreover, knockdown of CYLD enhanced mitogen-activated protein kinase (MAPK) ERK- and p38-mediated expression of c-jun, c-fos, and c-myc, which govern Nrf2 expression in cardiomyocytes. The CYLD deficiency-induced suppression of reactive oxygen species (ROS) formation, death and hypertrophy in cardiomyocytes was blocked by additional knockdown of Nrf2. Taken together, our findings demonstrate for the first time that CYLD mediates cardiac maladaptive remodeling and dysfunction, most likely via enhancing myocardial oxidative stress in response to pressure overload. At the molecular level, CYLD interrupts the ERK- and p38-/AP-1 and c-Myc pathways to suppress Nrf2-operated antioxidative capacity, thereby enhancing oxidative stress in the heart.


Subject(s)
Cardiomegaly/physiopathology , Cysteine Endopeptidases/metabolism , Down-Regulation , NF-E2-Related Factor 2/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ventricular Remodeling , Animals , Cardiomegaly/complications , Cardiomegaly/diagnostic imaging , Cardiomegaly/enzymology , Deubiquitinating Enzyme CYLD , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Gene Silencing , Heart Failure/complications , Heart Failure/diagnostic imaging , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Male , Mice, Knockout , Models, Biological , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress , Pressure , Proto-Oncogene Proteins c-myc/metabolism , Rats , Signal Transduction , Survival Analysis , Transcription Factor AP-1/metabolism , Ultrasonography , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Cell Tissue Res ; 362(1): 97-113, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26022334

ABSTRACT

Proliferative or synthetic vascular smooth muscle cells (VSMCs) are widely accepted to be mainly derived from the dedifferentiation or phenotypic modulation of mature contractile VSMCs, i.e., a phenotype switch from a normally quiescent and contractile type into a proliferative or synthetic form. However, this theory has been challenged by recent evidence that synthetic VSMCs predominantly originate instead from media-derived multipotent vascular stem cells (MVSCs). To test these hypotheses further, we re-examine whether the conventional rat aortic SMC (RASMC) culture involves the VSMC differentiation of MVSCs or the dedifferentiation of mature VSMCs and the potential mechanism for controlling the synthetic phenotype of RASMCs. We enzymatically isolated RASMCs and cultured the cells in both a regular growth medium (RGM) and a stem cell growth medium (SCGM). Regardless of culture conditions, only a small portion of freshly isolated RASMCs attaches, survives and grows slowly during the first 7 days of primary culture, while expressing both SMC- and MVSC-specific markers. RGM-cultured cells undergo a process of synthetic SMC differentiation, whereas SCGM-cultured cells can be differentiated into not only synthetic SMCs but also other somatic cells. Notably, compared with the RGM-cultured differentiated RASMCs, the SCGM-cultured undifferentiated cells exhibit the phenotype of MVSCs and generate greater amounts of reactive oxygen species (ROS) that act as a negative regulator of differentiation into synthetic VSMCs. Knockdown of phospholipase A2, group 7 (Pla2g7) suppresses ROS formation in the MVSCs while enhancing SMC differentiation of MVSCs. These results suggest that cultured synthetic VSMCs can be derived from the SMC differentiation of MVSCs with ROS as a negative regulator.


Subject(s)
Aorta/metabolism , Multipotent Stem Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Aorta/cytology , Cell Differentiation , Male , Multipotent Stem Cells/cytology , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reactive Oxygen Species
10.
J Ethnopharmacol ; 168: 326-36, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-25882312

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: American ginseng is capable of ameliorating cardiac dysfunction and activating Nrf2, a master regulator of antioxidant defense, in the heart. This study was designed to isolate compounds from American ginseng and to determine those responsible for the Nrf2-mediated resolution of inflamed macrophage-induced cardiomyocyte hypertrophy. MATERIALS AND METHODS: A standardized crude extract of American ginseng was supplied by the National Research Council of Canada, Institute for National Measurement Standards. A bioassay-based fractionization of American ginseng was performed to identify the putative substances which could activate Nrf2-mediated suppression of pro-inflammatory cytokine expression in macrophages and macrophage-mediated pro-hypertrophic growth in cardiomyocytes. RESULTS: A hexane fraction of an anti-inflammatory crude extract of American ginseng was found to be most effective in suppressing the inflammatory responses in macrophages. Preparative, reverse-phase HPLC and a comparative analysis by analytical scale LC-UV/MS revealed the hexane fraction contains predominantly C17 polyacetylenes and linolenic acid. Panaxynol, one of the major polyacetylenes, was found to be a potent Nrf2 activator. Panaxynol posttranscriptionally activated Nrf2 by inhibiting Kelch-like ECH-associated protein (Keap) 1-mediated degradation without affecting the binding of Keap1 and Nrf2. Moreover, panaxynol suppressed a selected set of cytokine expression via the activation of Nrf2 while minimally regulating nuclear factor-kappa B (NF-κB)-mediated cytokine expression in macrophages. It also dramatically inhibited the inflamed macrophage-mediated cardiomyocyte death and hypertrophy by activating Nrf2 in macrophages. CONCLUSIONS: These results demonstrate that American ginseng-derived panaxynol is a specific Nrf2 activator and panaxynol-activated Nrf2 signaling is at least partly responsible for American ginseng-induced health benefit in the heart.


Subject(s)
Diynes/pharmacology , Fatty Alcohols/pharmacology , Macrophages/drug effects , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/metabolism , Panax , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Cell Line , Cytokines/metabolism , Diynes/isolation & purification , Fatty Alcohols/isolation & purification , Female , Hypertrophy/drug therapy , Hypertrophy/metabolism , Macrophages/metabolism , Male , Mice, Knockout , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type II/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Rats
11.
Vascul Pharmacol ; 67-69: 59-66, 2015.
Article in English | MEDLINE | ID: mdl-25869499

ABSTRACT

Vascular smooth muscle cells (VSMCs) play a crucial role in atherosclerotic lesion formation. Sparstolonin B (SsnB) is a TLR2/TLR4 antagonist that inhibits inflammatory responses in multiple cell types. Herein, we investigated if SsnB inhibited VSMC proliferation, migration, inflammatory response and lipid accumulation. We found that SsnB suppressed VSMC proliferation and migration induced by PDGF. SsnB significantly suppressed the expression of MCP-1, TNFα and IL-6 in VSMCs stimulated by either lipopolysaccharide (LPS) or PDGF. Erk1/2 and Akt signaling pathways, which are responsible for the VSMC inflammatory response, were activated by LPS or PDGF stimulation, and SsnB significantly inhibited their activation. SsnB also substantially suppressed the intracellular cholesterol accumulation in VSMCs loaded with acetylated LDL. Mechanistically, SsnB remarkably repressed LPS-induced up-regulation of CD36, which is responsible for lipid uptake, and dramatically reversed LPS-induced inhibition of ABCA1, which promotes the efflux of intracellular free cholesterol. In conclusion, our results indicate that SsnB significantly inhibits VSMC proliferation, migration, inflammatory responses and lipid accumulation. Along with the previously reported anti-inflammatory activities of SsnB on macrophages and vascular endothelial cells, our data strongly suggest that SsnB may be developed as a new anti-atherogenic therapy.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammation Mediators/metabolism , Lipids/blood , Muscle, Smooth, Vascular/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Inflammation Mediators/antagonists & inhibitors , Lipids/antagonists & inhibitors , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Rats, Sprague-Dawley
12.
Heart Lung Circ ; 24(9): 919-24, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25837018

ABSTRACT

BACKGROUND: Angiotensin converting enzyme (ACE) inhibitors such as lisinopril, represent the front line pharmacological treatment for heart failure, which is characterised by marked left ventricular (LV) dilatation and hypertrophy. This study sought to determine whether initiating treatment with ACE inhibitors at different stages in the remodelling process would alter the efficacy of treatment. METHODS: To this end, LV size and function were determined in the aortocaval (AV) fistula model of volume overload-induced heart failure. Sprague-Dawley rats were assigned to sham, untreated AV fistula (21 weeks), AV fistula treated with lisinopril (21 weeks), or AV fistula treated with lisinopril from six to 21 weeks post-fistula groups. RESULTS: Administration of lisinopril for the entire 21-week period prevented LV dilatation, attenuated myocardial hypertrophy and prevented changes in myocardial compliance and contractility, whereas delaying initiation of treatment until six weeks post-fistula attenuated LV dilatation and hypertrophy, however, the delayed onset of treatment had no beneficial effect on ventricular compliance or systolic function. CONCLUSIONS: The results demonstrate differential effects that can occur with ACE inhibitors depending on the stage during the remodelling process at which treatment is administered.


Subject(s)
Cardiomegaly , Heart Failure , Lisinopril/pharmacology , Myocardial Contraction/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiomegaly/drug therapy , Cardiomegaly/physiopathology , Disease Models, Animal , Heart Failure/drug therapy , Heart Failure/physiopathology , Male , Rats , Rats, Sprague-Dawley
13.
Curr Drug Targets ; 16(4): 284-94, 2015.
Article in English | MEDLINE | ID: mdl-25342597

ABSTRACT

The conserved cylindromatosis (CYLD) codes for a deubiquitinating enzyme and is a crucial regulator of diverse cellular processes such as immune responses, inflammation, death, and proliferation. It directly regulates multiple key signaling cascades, such as the Nuclear Factor kappa B [NFkB] and the Mitogen-Activated Protein Kinase (MAPK) pathways, by its catalytic activity on polyubiquitinated key intermediates. Several lines of emerging evidence have linked CYLD to the pathogenesis of various maladies, including cancer, poor infection control, lung fibrosis, neural development, and now cardiovascular dysfunction. While CYLD-mediated signaling is cell type and stimuli specific, the activity of CYLD is tightly controlled by phosphorylation and other regulators such as Snail. This review explores a broad selection of current and past literature regarding CYLD's expression, function and regulation with emerging reports on its role in cardiovascular disease.


Subject(s)
Cardiovascular Diseases/enzymology , Cardiovascular System/enzymology , Inflammation Mediators/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Autophagy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular System/immunology , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Cell Cycle , Deubiquitinating Enzyme CYLD , Humans , Inflammation Mediators/immunology , Tumor Suppressor Proteins/immunology , Ubiquitin-Specific Proteases/immunology
14.
Methods Mol Biol ; 1220: 121-39, 2015.
Article in English | MEDLINE | ID: mdl-25388248

ABSTRACT

Cardiac mast cells store and release a variety of biologically active mediators, several of which have been implicated in the activation of matrix metalloproteinases in the volume-overloaded heart, while others are involved in the fibrotic process in pressure-overloaded hearts. Increased numbers of mast cells have been reported in explanted human hearts with dilated cardiomyopathy and in animal models of experimentally induced hypertension, myocardial infarction, and chronic cardiac volume overload. Also, there is evolving evidence implicating the cardiac mast cell as having a major role in the adverse remodeling underlying these cardiovascular disorders. Thus, the cardiac mast cell is the focus of this chapter that begins with a historical background, followed by sections on methods for their isolation and characterization, endogenous secretagogues, phenotype, and ability of estrogen to alter their phenotype so as to provide cardioprotection. Finally the role of mast cells in myocardial remodeling secondary to a sustained cardiac volume overload, hypertension, and ischemic injury and future research directions are discussed.


Subject(s)
Mast Cells/immunology , Myocardium/immunology , Myocardium/pathology , Ventricular Remodeling/immunology , Animals , Cell Separation , Heart Diseases/immunology , Heart Diseases/pathology , Humans , Mast Cells/cytology
15.
Hypertension ; 65(2): 328-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25403608

ABSTRACT

Estrogen regulation of myocardial chymase and chymase effects on cardiac remodeling are unknown. To test the hypothesis that estrogen prevents pressure overload-induced adverse cardiac remodeling by inhibiting mast cell (MC) chymase release, transverse aortic constriction or sham surgery was performed in 7-week-old intact and ovariectomized (OVX) rats. Three days before creating the constriction, additional groups of OVX rats began receiving 17ß-estradiol, a chymase inhibitor, or a MC stabilizer. Left ventricular function, cardiomyocyte size, collagen volume fraction, MC density and degranulation, and myocardial and plasma chymase levels were assessed 18 days postsurgery. Aortic constriction resulted in ventricular hypertrophy in intact and OVX groups, whereas collagen volume fraction was increased only in OVX rats. Chymase protein content was increased by aortic constriction in the intact and OVX groups, with the magnitude of the increase being greater in OVX rats. MC density and degranulation, plasma chymase levels, and myocardial active transforming growth factor-ß1 levels were increased by aortic constriction only in OVX rats. Estrogen replacement markedly attenuated the constriction-increased myocardial chymase, MC density and degranulation, plasma chymase, and myocardial active transforming growth factor-ß1, as well as prevented ventricular hypertrophy and increased collagen volume fraction. Chymostatin attenuated the aortic constriction-induced ventricular hypertrophy and collagen volume fraction in the OVX rats similar to that achieved by estrogen replacement. Nedocromil yielded similar effects, except for the reduction of chymase content. We conclude that the estrogen-inhibited release of MC chymase is responsible for the cardioprotection against transverse aortic constriction-induced adverse cardiac remodeling.


Subject(s)
Chymases/metabolism , Estradiol/pharmacology , Hypertrophy, Left Ventricular/prevention & control , Mast Cells/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiomyopathy, Hypertrophic/complications , Cell Degranulation/drug effects , Cell Size/drug effects , Collagen/analysis , Estrogen Replacement Therapy , Female , Hypertrophy, Left Ventricular/etiology , Mast Cells/enzymology , Mast Cells/metabolism , Mast Cells/physiology , Myocardium/enzymology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Nedocromil/pharmacology , Oligopeptides/pharmacology , Organ Size/drug effects , Ovariectomy , Random Allocation , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/prevention & control
16.
Cardiovasc Drugs Ther ; 28(5): 433-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25117676

ABSTRACT

PURPOSE: Ischemia/reperfusion results in tissue damage, a rapid increase in cytokines and chemokines and inflammatory cell infiltration. Herein we investigated the ability of a selective TLR2/4 antagonist, Sparstolonin B (SsnB), to protect rat cultured left ventricular tissue (LV) slices from hypoxic injury by inhibiting the myocardial inflammatory response independent of inflammatory cell infiltration. METHODS AND RESULTS: Media Lactate dehydrogenase (LDH) levels were measured to reflect hypoxia-induced cytotoxicity and cell injury with and without SsnB. Incubation with SsnB (15 and 30 µM) significantly reduced by 20 and 40%, respectively, the amount of LDH released from the hypoxic LV slices. TUNEL staining showed that SsnB significantly attenuated the levels of hypoxia-induced apoptotic cells from 61.5 ± 4.0 to 27.0 ± 2.1 (15 µM SsnB) and 23.5 ± 2.2 (30 µM SsnB) cells/unit area. Similarly, the Periodic Acid-Schiff (PAS) staining of ischemic areas in untreated hypoxic LV slices was increased 17 fold from 0.26± 0.09 to 4.41 ± 0.43%, while in hypoxic slices incubated with 15 and 30 µM of SsnB, the PAS positive ischemic areas were increased by only 6.4 fold to 1.66 ± 0.39% and 3.8 fold to 1.00 ± 0.22%, respectively. Rt-PCR confirmed that MCP1 and IL-6 expression during hypoxia was elevated by 2 and 4 fold, respectively, while their up-regulation was significantly inhibited (i.e., < 0.7 fold increase) by SsnB. CONCLUSION: The selective TLR2/4 antagonist, Sparstolonin B, can substantially protect LV myocardium via its ability to inhibit injury resulting from hypoxic myocardial-generated inflammation. Accordingly SsnB has potential as a therapeutic agent for the attenuation of myocardial ischemia-reperfusion injury.


Subject(s)
Apoptosis/drug effects , Heart Ventricles/pathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Hypoxia/drug therapy , Inflammation/drug therapy , Necrosis/drug therapy , Animals , Chemokine CCL2/biosynthesis , Dose-Response Relationship, Drug , Heart Ventricles/drug effects , Hypoxia/complications , Interleukin-6/biosynthesis , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Reperfusion Injury/drug therapy , Necrosis/complications , Rats , Tissue Culture Techniques , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Up-Regulation/drug effects
17.
Oxid Med Cell Longev ; 2014: 748524, 2014.
Article in English | MEDLINE | ID: mdl-24895528

ABSTRACT

The anticancer therapy of doxorubicin (Dox) has been limited by its acute and chronic cardiotoxicity. In addition to a causative role of oxidative stress, autophagy appears to play an important role in the regulation of Dox-induced cardiotoxicity. However, the underlying mechanisms remain unclear. Accordingly, we explored a role of nuclear factor erythroid-2 related factor 2 (Nrf2) in Dox-induced cardiomyopathy with a focus on myocardial oxidative stress and autophagic activity. In wild type (WT) mice, a single intraperitoneal injection of 25 mg/kg Dox rapidly induced cardiomyocyte necrosis and cardiac dysfunction, which were associated with oxidative stress, impaired autophagy, and accumulated polyubiquitinated protein aggregates. However, these Dox-induced adverse effects were exaggerated in Nrf2 knockout (Nrf2(-/-)) mice. In cultured cardiomyocytes, overexpression of Nrf2 increased the steady levels of LC3-II, ameliorated Dox-induced impairment of autophagic flux and accumulation of ubiquitinated protein aggregates, and suppressed Dox-induced cytotoxicity, whereas knockdown of Nrf2 exerted opposite effects. Moreover, the exaggerated adverse effects in Dox-intoxicated Nrf2 depleted cardiomyocytes were dramatically attenuated by forced activation of autophagy via overexpression of autophagy related gene 5 (Atg5). Thus, these results suggest that Nrf2 is likely an endogenous suppressor of Dox-induced cardiotoxicity by controlling both oxidative stress and autophagy in the heart.


Subject(s)
Doxorubicin/toxicity , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiotoxicity , Cells, Cultured , Heart/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Necrosis , Oxidative Stress/drug effects
18.
FEBS Lett ; 588(12): 2115-24, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24815692

ABSTRACT

Nrf2 appears to be a critical regulator of diabetes in rodents. However, the underlying mechanisms as well as the clinical relevance of the Nrf2 signaling in human diabetes remain to be fully understood. Herein, we report that islet expression of Nrf2 is upregulated at an earlier stage of diabetes in both human and mice. Activation of Nrf2 suppresses oxidative stress and oxidative stress-induced ß-cell apoptosis while enhancing autophagic clearance in isolated rat islets. Additionally, oxidative stress per se activated autophagy in ß-cells. Thus, these results reveal that Nrf2 drives a novel antioxidant independent autophagic clearance for ß-cell protection in the setting of diabetes.


Subject(s)
Autophagy/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/analogs & derivatives , Oxidative Stress/drug effects , Animals , Cell Survival/drug effects , Humans , Hydrogen Peroxide/pharmacology , Insulin-Secreting Cells/metabolism , Male , Mice , Middle Aged , Oleanolic Acid/pharmacology , Rats , Ubiquitination/drug effects
19.
J Mol Cell Cardiol ; 72: 305-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24747945

ABSTRACT

Nuclear factor erythroid-2 related factor 2 (Nrf2) is a master transcription factor that controls the basal and inducible expression of a battery of antioxidant genes and other cytoprotective phase II detoxifying enzymes. While knockout of Nrf2 exaggerates cardiac pathological remodeling and dysfunction in diverse pathological settings, pharmacological activation of Nrf2 protects against cardiomyocyte injury and cardiac dysfunction. In contrast, there is also a concern that the chronic activation of Nrf2 secondary to oxidative stress is a contributing mechanism for the reductive stress-mediated heart failure. However, a direct link between cardiac specific activation of Nrf2 and cardiac protection or dysfunction in vivo remains to be established. Therefore, we investigated the effect of cardiomyocyte-specific transgenic activation of Nrf2 (Nrf2(ctg)) on cardiac pathological remodeling and dysfunction. We found that the cardiomyocyte-specific activation of Nrf2 suppressed myocardial oxidative stress as well as cardiac apoptosis, fibrosis, hypertrophy, and dysfunction in a setting of sustained pressure overload induced by transverse aortic arch constriction (TAC) in mice. Notably, the constitutive activation of Nrf2 increased the steady level of autophagosomes while decreasing the ubiquitinated protein aggregates in the heart after TAC. Nrf2 gene gain- and loss-of-function approaches revealed that Nrf2 enhances autophagosome formation and autophagic flux in cardiomyocytes. Unexpectedly, while Nrf2 minimally regulated apoptosis, it suppressed significantly the proteotoxic necrosis in cardiomyocytes. In addition, Nrf2 attenuated the proteocytotoxicity presumably via enhancing autophagy-mediated clearance of ubiquitinated protein aggregates in cardiomyocytes. Taken together, we demonstrated for the first time that cardiac specific activation of Nrf2 suppresses cardiac maladaptive remodeling and dysfunction most likely by enhancing autophagic clearance of toxic protein aggregates in the heart.


Subject(s)
Autophagy/genetics , Cardiomegaly/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , NF-E2-Related Factor 2/genetics , Ubiquitinated Proteins/metabolism , Animals , Apoptosis , Cardiomegaly/metabolism , Cardiomegaly/pathology , Fibrosis , Gene Expression , Male , Mice , Mice, Transgenic , Myocardium/pathology , Myocytes, Cardiac/pathology , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Protein Aggregation, Pathological , Proteolysis , Rats , Signal Transduction , Ubiquitin/metabolism
20.
Biochem Biophys Res Commun ; 444(4): 555-61, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24486487

ABSTRACT

Nuclear factor erythroid 2-related factor (Nrf2) is the major regulator of cellular defenses against various pathological stresses in a variety of organ systems, thus Nrf2 has evolved to be an attractive drug target for the treatment and/or prevention of human disease. Several synthetic oleanolic triterpenoids including dihydro-CDDO-trifluoroethyl amide (dh404) appear to be potent activators of Nrf2 and exhibit chemopreventive promises in multiple disease models. While the pharmacological efficacy of Nrf2 activators may be dependent on the nature of Nrf2 activation in specific cell types of target organs, the precise role of Nrf2 in mediating biological effects of Nrf2 activating compounds in various cell types remains to be further explored. Herein we report a unique and Nrf2-dependent anti-inflammatory profile of dh404 in inflamed macrophages. In lipopolysaccharide (LPS)-inflamed RAW264.7 macrophages, dh404 dramatically suppressed the expression of pro-inflammatory cytokines including inducible nitric oxide synthase (iNOS), monocyte chemotactic protein-1 (MCP-1), and macrophage inflammatory protein-1 beta (MIP-1ß), while minimally regulating the expression of interleulin-6 (IL-6), IL-1ß, and tumor necrosis factor alpha (TNFα). Dh404 potently activated Nrf2 signaling; however, it did not affect LPS-induced NF-κB activity. Dh404 did not interrupt the interaction of Nrf2 with its endogenous inhibitor Kelch-like ECH associating protein 1 (Keap1) in macrophages. Moreover, knockout of Nrf2 blocked the dh404-induced anti-inflammatory responses in LPS-inflamed macrophages. These results demonstrated that dh404 suppresses pro-inflammatory responses in macrophages via an activation of Nrf2 independently of Keap1 and NF-κB, suggesting a unique therapeutic potential of dh404 for specific targeting a Nrf2-mediated resolution of inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Macrophages/immunology , NF-E2-Related Factor 2/immunology , Oleanolic Acid/analogs & derivatives , Animals , Cell Line , Cells, Cultured , Chemokine CCL2/immunology , Chemokine CCL4/immunology , Cytokines/immunology , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-kappa B/immunology , Nitric Oxide Synthase Type II/immunology , Oleanolic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...