Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Circulation ; 142(8): 758-775, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32489148

ABSTRACT

BACKGROUND: Cardiac rupture is a major lethal complication of acute myocardial infarction (MI). Despite significant advances in reperfusion strategies, mortality from cardiac rupture remains high. Studies suggest that cardiac rupture can be accelerated by thrombolytic therapy, but the relevance of this risk factor remains controversial. METHODS: We analyzed protease-activated receptor 4 (Par4) expression in mouse hearts with MI and investigated the effects of Par4 deletion on cardiac remodeling and function after MI by echocardiography, quantitative immunohistochemistry, and flow cytometry. RESULTS: Par4 mRNA and protein levels were increased in mouse hearts after MI and in isolated cardiomyocytes in response to hypertrophic and inflammatory stimuli. Par4-deficient mice showed less myocyte apoptosis, reduced infarct size, and improved functional recovery after acute MI relative to wild-type (WT). Conversely, Par4-/- mice showed impaired cardiac function, greater rates of myocardial rupture, and increased mortality after chronic MI relative to WT. Pathological evaluation of hearts from Par4-/- mice demonstrated a greater infarct expansion, increased cardiac hemorrhage, and delayed neutrophil accumulation, which resulted in impaired post-MI healing compared with WT. Par4 deficiency also attenuated neutrophil apoptosis in vitro and after MI in vivo and impaired inflammation resolution in infarcted myocardium. Transfer of Par4-/- neutrophils, but not of Par4-/- platelets, in WT recipient mice delayed inflammation resolution, increased cardiac hemorrhage, and enhanced cardiac dysfunction. In parallel, adoptive transfer of WT neutrophils into Par4-/- mice restored inflammation resolution, reduced cardiac rupture incidence, and improved cardiac function after MI. CONCLUSIONS: These findings reveal essential roles of Par4 in neutrophil apoptosis and inflammation resolution during myocardial healing and point to Par4 inhibition as a potential therapy that should be limited to the acute phases of ischemic insult and avoided for long-term treatment after MI.


Subject(s)
Gene Expression Regulation , Heart Rupture , Myocardial Infarction , Myocardium/metabolism , Receptors, Thrombin/deficiency , Animals , Female , Heart Rupture/etiology , Heart Rupture/genetics , Heart Rupture/metabolism , Heart Rupture/prevention & control , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Knockout , Myocardial Infarction/classification , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Receptors, Thrombin/biosynthesis
2.
JACC Basic Transl Sci ; 5(1): 69-83, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32043021

ABSTRACT

Activated factor X is a key component of the coagulation cascade, but whether it directly regulates pathological cardiac remodeling is unclear. In mice subjected to pressure overload stress, cardiac factor X mRNA expression and activity increased concurrently with cardiac hypertrophy, fibrosis, inflammation and diastolic dysfunction, and responses blocked with a low coagulation-independent dose of rivaroxaban. In vitro, neurohormone stressors increased activated factor X expression in both cardiac myocytes and fibroblasts, resulting in activated factor X-mediated activation of protease-activated receptors and pro-hypertrophic and -fibrotic responses, respectively. Thus, inhibition of cardiac-expressed activated factor X could provide an effective therapy for the prevention of adverse cardiac remodeling in hypertensive patients.

3.
Circ Heart Fail ; 13(1): e006426, 2020 01.
Article in English | MEDLINE | ID: mdl-31916447

ABSTRACT

BACKGROUND: The failing right ventricle (RV) does not respond like the left ventricle (LV) to guideline-directed medical therapy of heart failure, perhaps due to interventricular differences in their molecular pathophysiology. METHODS: Using the canine tachypacing-induced biventricular heart failure (HF) model, we tested the hypothesis that interventricular differences in microRNAs (miRs) expression distinguish failing RV from failing LV. RESULTS: Severe RV dysfunction was indicated by elevated end-diastolic pressure (11.3±2.5 versus 5.7±2.0 mm Hg; P<0.0001) and diminished fractional area change (24.9±7.1 versus 48.0±3.6%; P<0.0001) relative to prepacing baselines. Microarray analysis of ventricular tissue revealed that miR-21 and miR-221, 2 activators of profibrotic and proliferative processes, increased the most, at 4- and 2-fold, respectively, in RV-HF versus RV-Control. Neither miR-21 or miR-221 was statistically significantly different in LV-HF versus LV-Control. These changes were accompanied by more extensive fibrosis in RV-HF than LV-HF. To test whether miR-21 and miR-221 upregulation is specific to RV cellular response to mechanical and hormonal stimuli associated with HF, we subjected fibroblasts and cardiomyocytes isolated from normal canine RV and LV to cyclic overstretch and aldosterone. These 2 stressors markedly upregulated miR-21 and miR-221 in RV fibroblasts but not in LV fibroblasts nor cardiomyocytes of either ventricle. Furthermore, miR-21/221 knockdown significantly attenuated RV but not LV fibroblast proliferation. CONCLUSIONS: We identified a novel, biological difference between RV and LV fibroblasts that might underlie distinctions in pathological remodeling of the RV in biventricular HF.


Subject(s)
Fibroblasts/metabolism , Heart Failure/metabolism , Heart Ventricles/metabolism , MicroRNAs/metabolism , Ventricular Dysfunction, Right/metabolism , Animals , Dogs , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Myocytes, Cardiac/metabolism , Up-Regulation , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Left/physiology
4.
Cell Physiol Biochem ; 53(6): 982-998, 2019.
Article in English | MEDLINE | ID: mdl-31829530

ABSTRACT

BACKGROUND/AIMS: Diabetic cardiomyopathy (DCM) is characterized by structural and functional alterations that can lead to heart failure. Several mechanisms are known to be involved in the pathogenesis of DCM, however, the molecular mechanism that links inflammation to DCM is incompletely understood. To learn about this mechanism, we investigated the role of inflammatory serine proteases (ISPs) during the development of DCM. METHODS: Eight weeks old mice with deletion of dipeptidyl peptidase I (DPPI), an enzyme involved in the maturation of major ISPs, and wild type (WT) mice controls were injected with streptozotocin (50 mg/kg for 5 days intraperitoneally) and studied after 4, 8, 16, and 20 week after induction of type 1 diabetes mellitus (T1DM). Induction of diabetes was followed by echocardiographic measurements, glycemic and hemoglobulin A1c profiling, immunoblot, qPCR, enzyme activity assays, and immunohistochemistry (IHC) analysis of DPPI, ISPs, and inflammatory markers. Fibrosis was determined from left ventricular heart by Serius Red staining and qPCR. Apoptosis was determined by TUNEL assay and immunoblot analysis. RESULTS: In the diabetic WT mice, DPPI expression increased along with ISP activation, and DPPI accumulated abundantly in the left ventricle mainly from infiltrating neutrophils. In diabetic DPPI-knockout (DPPI-KO) mice, significantly decreased activation of ISPs, myocyte apoptosis, fibrosis, and cardiac function was improved compared to diabetic WT mice. In addition, DPPI-KO mice showed a decrease in overall inflammatory status mediated by diabetes induction which was manifested by decreased production of pro-inflammatory cytokines like TNF-α, IL-1ß and IL-6. CONCLUSION: This study elucidates a novel role of ISPs in potentiating the immunological responses that lead to the pathogenesis of DCM in T1DM. To the best of our knowledge, this is the first study to report that DPPI expression and activation promotes the inflammation that enhances myocyte apoptosis and contributes to the adverse cardiac remodeling that subsequently leads to DCM.


Subject(s)
Cathepsin C/metabolism , Diabetic Cardiomyopathies/pathology , Serine Proteases/metabolism , Animals , Apoptosis , Blood Glucose/analysis , Cathepsin C/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/etiology , Down-Regulation , Fibrosis , Heart/physiology , Heart Ventricles/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism
5.
J Mol Cell Cardiol ; 134: 29-39, 2019 09.
Article in English | MEDLINE | ID: mdl-31252040

ABSTRACT

BACKGROUND: Inflammatory serine proteases (ISPs) play an important role in cardiac repair after injury through hydrolysis of dead cells and extracellular matrix (ECM) debris. Evidence also suggests an important role of ISPs in the coordination of the inflammatory response. However, the effect of ISPs on inflammation is obfuscated by the confounding factors associated with cell death and inflammatory cell infiltration induced after cardiac injury. This study investigated whether neutrophil-derived cathepsin G (Cat.G) influences inflammation and remodeling in the absence of prior cardiac injury and cell death. METHODS AND RESULTS: Intracardiac catheter delivery of Cat.G (1 mg/kg) in rats induced significant left ventricular (LV) dilatation and cardiac contractile dysfunction at day 5, but not at day 2, post-delivery compared to vehicle-treated animals. Cat.G delivery also significantly increased matrix metalloprotease activity and collagen and fibronectin degradation at day 5 compared to vehicle-treated rats and these changes were associated with increased death signaling pathways and myocyte apoptosis. Mechanistic analysis shows that Cat.G-treatment induced potent chemotactic activity in hearts at day 2 and 5 post-delivery, characterized by processing and activation of interleukin (IL)-1ß and IL-18, stimulation of inflammatory signaling pathways and accumulation of myeloid cells when compared to vehicle-treated rats. Cat.G-induced processing of IL-1ß and IL-18 was independent of the canonical NLRP-3 inflammasome pathway and treatment of isolated cardiomyocytes with inhibitors of NLRP-3 or caspase-1 failed to reduce Cat.G-induced cardiomyocyte death. Notably, rats treated with IL-1 receptor antagonist (IL-1Ra) show reduced inflammation and improved cardiac remodeling and function following Cat.G delivery. CONCLUSIONS: Cat.G exerts potent chemoattractant and pro-inflammatory effects in non-stressed or injured heart in part through processing and activation of IL-1 family cytokines, subsequently leading to adverse cardiac remodeling and function. Thus, targeting ISPs could be a novel therapeutic strategy to reduce cardiac inflammation and improve cardiac remodeling and function after injury or stress.


Subject(s)
Atrial Remodeling/drug effects , Cardiac Catheters , Cathepsin G/administration & dosage , Inflammasomes/drug effects , Inflammation/chemically induced , Ventricular Remodeling/drug effects , Animals , Cardiac Catheterization , Cathepsin G/adverse effects , Cathepsin G/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Male , Neutrophils/enzymology , Neutrophils/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
6.
Basic Res Cardiol ; 112(6): 62, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28913553

ABSTRACT

Early reperfusion of ischemic cardiac tissue increases inflammatory cell infiltration which contributes to cardiomyocyte death and loss of cardiac function, referred to as ischemia/reperfusion (IR) injury. Neutrophil- and mast cell-derived proteases, cathepsin G (Cat.G) and chymase, are released early after IR, but their function is complicated by potentially redundant actions and targets. This study investigated whether a dual inhibition of Cat.G and chymase influences cardiomyocyte injury and wound healing after experimental IR in mice. Treatment with a dual Cat.G and chymase inhibitor (DCCI) immediately after reperfusion blocked cardiac Cat.G and chymase activity induced after IR, which resulted in decreased immune response in the infarcted heart. Mice treated with DCCI had less myocardial collagen deposition and showed preserved ventricular function at 1 and 7 days post-IR compared with vehicle-treated mice. DCCI treatment also significantly attenuated focal adhesion (FA) complex disruption and myocyte degeneration after IR. Treatment of isolated cardiomyocytes with Cat.G or chymase significantly promoted FA signaling downregulation, myofibril degeneration and myocyte apoptosis. Conversely, treatment of cardiac fibroblasts with Cat.G or chymase induced FA signaling activation and increased their migration and differentiation to myofibroblasts. These opposite responses in cardiomyocytes and fibroblasts were blocked by treatment with DCCI. These findings show that Cat.G and chymase are key mediators of myocyte apoptosis and fibroblast migration and differentiation that play a role in adverse cardiac remodeling and function post-IR. Thus, dual targeting of neutrophil- and mast cell-derived proteases could be used as a novel therapeutic strategy to reduce post-IR inflammation and improve cardiac remodeling.


Subject(s)
Atrial Remodeling/physiology , Cathepsin G/antagonists & inhibitors , Chymases/antagonists & inhibitors , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/pathology , Animals , Apoptosis/physiology , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology
7.
J Mol Cell Cardiol ; 90: 21-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26643815

ABSTRACT

Protease-activated receptor (PAR)4 is a low affinity thrombin receptor with less understood function relative to PAR1. PAR4 is involved in platelet activation and hemostasis, but its specific actions on myocyte growth and cardiac function remain unknown. This study examined the role of PAR4 deficiency on cardioprotection after myocardial ischemia-reperfusion (IR) injury in mice. When challenged by in vivo or ex vivo IR, PAR4 knockout (KO) mice exhibited increased tolerance to injury, which was manifest as reduced infarct size and a more robust functional recovery compared to wild-type mice. PAR4 KO mice also showed reduced cardiomyocyte apoptosis and putative signaling shifts in survival pathways in response to IR. Inhibition of PAR4 expression in isolated cardiomyocytes by shRNA offered protection against thrombin and PAR4-agonist peptide-induced apoptosis, while overexpression of wild-type PAR4 significantly enhanced the susceptibility of cardiomyocytes to apoptosis, even under low thrombin concentrations. Further studies implicate Src- and epidermal growth factor receptor-dependent activation of JNK on the proapoptotic effect of PAR4 in cardiomyocytes. These findings reveal a pivotal role for PAR4 as a regulator of cardiomyocyte survival and point to PAR4 inhibition as a therapeutic target offering cardioprotection after acute IR injury.


Subject(s)
Myocardial Reperfusion Injury/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Receptors, Thrombin/genetics , Animals , Apoptosis/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Peptides/pharmacology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Thrombin/agonists , Receptors, Thrombin/antagonists & inhibitors , Receptors, Thrombin/deficiency , Signal Transduction , Thrombin/pharmacology , src-Family Kinases/genetics , src-Family Kinases/metabolism
8.
Circ Res ; 117(11): 926-32, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26383970

ABSTRACT

RATIONALE: Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-ß super family of secreted factors. A recent study showed that reduced GDF11 blood levels with aging was associated with pathological cardiac hypertrophy (PCH) and restoring GDF11 to normal levels in old mice rescued PCH. OBJECTIVE: To determine whether and by what mechanism GDF11 rescues aging dependent PCH. METHODS AND RESULTS: Twenty-four-month-old C57BL/6 mice were given a daily injection of either recombinant (r) GDF11 at 0.1 mg/kg or vehicle for 28 days. rGDF11 bioactivity was confirmed in vitro. After treatment, rGDF11 levels were significantly increased, but there was no significant effect on either heart weight or body weight. Heart weight/body weight ratios of old mice were not different from 8- or 12-week-old animals, and the PCH marker atrial natriuretic peptide was not different in young versus old mice. Ejection fraction, internal ventricular dimension, and septal wall thickness were not significantly different between rGDF11 and vehicle-treated animals at baseline and remained unchanged at 1, 2, and 4 weeks of treatment. There was no difference in myocyte cross-sectional area rGDF11 versus vehicle-treated old animals. In vitro studies using phenylephrine-treated neonatal rat ventricular myocytes, to explore the putative antihypertrophic effects of GDF11, showed that GDF11 did not reduce neonatal rat ventricular myocytes hypertrophy, but instead induced hypertrophy. CONCLUSIONS: Our studies show that there is no age-related PCH in disease-free 24-month-old C57BL/6 mice and that restoring GDF11 in old mice has no effect on cardiac structure or function.


Subject(s)
Aging/pathology , Bone Morphogenetic Proteins/pharmacology , Cardiomegaly/prevention & control , Growth Differentiation Factors/pharmacology , Myocytes, Cardiac/drug effects , Ventricular Remodeling/drug effects , Adrenergic alpha-1 Receptor Agonists/pharmacology , Age Factors , Aging/metabolism , Animals , Bone Morphogenetic Proteins/administration & dosage , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Drug Administration Schedule , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Growth Differentiation Factors/administration & dosage , Injections, Intraperitoneal , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recombinant Proteins/pharmacology , Time Factors , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
9.
Circulation ; 129(20): 2031-43, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24583314

ABSTRACT

BACKGROUND: The proto-oncogene Casitas b-lineage lymphoma (c-Cbl) is an adaptor protein with an intrinsic E3 ubiquitin ligase activity that targets receptor and nonreceptor tyrosine kinases, resulting in their ubiquitination and downregulation. However, the function of c-Cbl in the control of cardiac function is currently unknown. In this study, we examined the role of c-Cbl in myocyte death and cardiac function after myocardial ischemia. METHODS AND RESULTS: We show increased c-Cbl expression in human ischemic and dilated cardiomyopathy hearts and in response to pathological stress stimuli in mice. c-Cbl-deficient mice demonstrated a more robust functional recovery after myocardial ischemia/reperfusion injury and significantly reduced myocyte apoptosis and improved cardiac function. Ubiquitination and downregulation of key survival c-Cbl targets, epidermal growth factor receptors and focal adhesion kinase, were significantly reduced in c-Cbl knockout mice. Inhibition of c-Cbl expression or its ubiquitin ligase activity in cardiac myocytes offered protection against H2O2 stress. Interestingly, c-Cbl deletion reduced the risk of death and increased cardiac functional recovery after chronic myocardial ischemia. This beneficial effect of c-Cbl deletion was associated with enhanced neoangiogenesis and increased expression of vascular endothelial growth factor-a and vascular endothelial growth factor receptor type 2 in the infarcted region. CONCLUSIONS: c-Cbl activation promotes myocyte apoptosis, inhibits angiogenesis, and causes adverse cardiac remodeling after myocardial infarction. These findings point to c-Cbl as a potential therapeutic target for the maintenance of cardiac function and remodeling after myocardial ischemia.


Subject(s)
Cardiomyopathy, Dilated/physiopathology , Heart Failure/physiopathology , Myocardial Ischemia/physiopathology , Proto-Oncogene Proteins c-cbl/physiology , Adult , Aged , Animals , Apoptosis/physiology , Cardiac Catheterization , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Echocardiography , Electrocardiography , Female , Heart Failure/genetics , Heart Failure/pathology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-cbl/genetics , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/metabolism
10.
J Mol Cell Cardiol ; 53(2): 240-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22609523

ABSTRACT

Numerous studies demonstrated increased expression of extracellular matrix (ECM) proteins and activation of focal adhesion (FA) signaling pathways in models of pressure overload-induced cardiac hypertrophy. However, little is known about FA signaling in response to volume overload where cardiac hypertrophy is associated with ECM loss. This study examines the role of beta1-adrenergic receptors (ß(1)-ARs) in FA signaling changes and myocyte apoptosis induced during acute hemodynamic stress of volume overload. Rats with eccentric cardiac hypertrophy induced after aorto-caval fistula (ACF) develop reduced interstitial collagen content and decreased tyrosine phosphorylation of key FA signaling molecules FAK, Pyk(2) and paxillin along with an increase in cardiac myocyte apoptosis. ACF also increased activation of PTEN, a dual lipid and protein phosphatase, and its interaction with FA proteins. ß(1)-AR blockade (extended-release of metoprolol succinate, 100mg QD) markedly attenuated PTEN activation, restored FA signaling and reduced myocyte apoptosis induced by ACF at 2days, but failed to reduce interstitial collagen loss and left ventricular dilatation. Treating cultured myocytes with ß(1)-AR agonists or adenoviral expression of ß(1)-ARs caused PTEN activation and interaction with FA proteins, thus leading to FA signaling downregulation and myocyte apoptosis. Adenoviral-mediated expression of a catalytically inactive PTEN mutant or wild-type FAK restored FA signaling downregulation and attenuated myocyte apoptosis induced by ß(1)-ARs. Collectively, these data show that ß(1)-AR stimulation in response to ACF induces FA signaling downregulation through an ECM-independent mechanism. This effect involves PTEN activation and may contribute to adverse cardiac remodeling and function in the course of volume overload.


Subject(s)
Focal Adhesions/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-1/metabolism , Adrenergic Antagonists/pharmacology , Animals , Apoptosis/physiology , Arterio-Arterial Fistula/metabolism , Blotting, Western , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cells, Cultured , Immunoprecipitation , Male , PTEN Phosphohydrolase/metabolism , Pulmonary Artery/abnormalities , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/genetics , Signal Transduction/genetics , Signal Transduction/physiology
11.
J Biol Chem ; 287(8): 5327-39, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22203672

ABSTRACT

The neutrophil-derived serine protease, cathepsin G (Cat.G), has been shown to induce myocyte detachment and apoptosis by anoikis through down-regulation of focal adhesion (FA) signaling. However, the mechanisms that control FA protein stability and turnover in myocytes are not well understood. Here, we have shown that the Casitas b-lineage lymphoma (c-Cbl), adaptor protein with an intrinsic E3 ubiquitin ligase activity, is involved in FA and myofibrillar protein stability and turnover in myocytes. Cat.G treatment induced c-Cbl activation and its interaction with FA proteins. Deletion of c-Cbl using c-Cbl knock-out derived myocytes or inhibition of c-Cbl ligase activity significantly reduced FA protein degradation, myofibrillar degeneration, and myocyte apoptosis induced by Cat.G. We also found that inhibition of the proteasome activity, but not the lysosome or the calpain activity, markedly attenuated FA and myofibrillar protein degradation induced by Cat.G. Interestingly, c-Cbl activation induced by Cat.G was mediated through epidermal growth factor receptor (EGFR) transactivation as inhibition of EGFR kinase activity markedly attenuated c-Cbl phosphorylation and FA protein degradation induced by Cat.G. These findings support a model in which neutrophil protease Cat.G promotes c-Cbl interaction with FA proteins, resulting in enhanced c-Cbl-mediated FA protein ubiquitination and degradation, myofibril degradation, and subsequent down-regulation of myocyte survival signaling.


Subject(s)
Cathepsin G/pharmacology , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Myofibrils/drug effects , Myofibrils/metabolism , Neutrophils/enzymology , Proto-Oncogene Proteins c-cbl/metabolism , Animals , Cell Survival/drug effects , Enzyme Activation/drug effects , Genes, erbB-1/genetics , Heart Ventricles/cytology , Heart Ventricles/injuries , Mice , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Rats , Signal Transduction/drug effects , Transcriptional Activation/drug effects
12.
Protein Expr Purif ; 72(1): 8-18, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20226862

ABSTRACT

An advanced protocol is provided to adapt cells for enhanced proliferation in and expression from deuterated minimal media. For large proteins (>20-30 kDa), deuteration levels >90% are essential for NMR characterization of structure and dynamics. In addition, the low sensitivity of NMR demands can be achieved without major sacrifice to yield. We applied the approach to human adult hemoglobin (Hb A), a 64 kDa, tetrameric protein that requires significant post-expression processing. This aspect accentuates the need for high yield. Using specially adapted JM109(DE3) Escherichia coli, we developed a shake-flask approach to express >90% deuterated NMR samples. Typical yields were 2.5-fold higher than obtained from cells adapted by more-traditional methods, while deuteration levels were increased by 17%. Ultimately, a 200 mL culture was sufficient to obtain ((2)H, (15)N)-labeled Hb A sufficient for a 200 microM, 400 microL NMR sample. This avoids need for additional equipment for fermentation, which was used in previous protocols to express Hb A. It also allows a much smaller culture volume than often required by such equipment, for corresponding linear reductions in the cost of labeled starting materials. We tested the adaptation protocol with both JM109 and JM109(DE3) E. coli, and with pre- and post-transformation with the Hb A expression plasmid (pHE7). The (DE3) strain consistently outperformed its parent strain in response to adaptation, with the latter failing to survive adaptation in multiple trials. In addition, pre-transformed cells were consistently more receptive to adaptation. Finally, we also detail updated protocols to isolate Hb A in its functional form.


Subject(s)
Escherichia coli/genetics , Hemoglobin A/chemistry , Hemoglobin A/genetics , Nuclear Magnetic Resonance, Biomolecular , Deuterium/metabolism , Gene Expression , Hemoglobin A/isolation & purification , Hemoglobin A/metabolism , Humans , Plasmids/genetics , Protein Conformation , Transformation, Genetic
13.
Article in English | MEDLINE | ID: mdl-16519407

ABSTRACT

Carboxyl modified PEG spacer was synthesized and linked covalently to cellulose beads. L-lysine ligand was coupled to the spacer and its selective affinity for low-density lipoprotein-cholesterol (LDL-C) was determined. It was found that the adsorption capacity and the efficiency of the ligand for adsorption of LDL-C was increased when PEG spacer was used. Experimental results showed that with the chain length of PEG spacers increased from 1000Da to 6000 Da, the average adsorption capacity of LDL-C was enhanced from 0.242 mg/ml to 0.903 mg/ml. Above results indicate that PEG spacers are conducible to the selective removal of LDL-C from human plasma. In addition, LDL-C adsorbent with PEG spacers has a low adsorption capacity for high-density lipoprotein (HDL-C).


Subject(s)
Blood Component Removal , Cellulose/chemistry , Cholesterol, LDL/chemistry , Lysine/chemistry , Polyethylene Glycols/chemistry , Adsorption , Blood Component Removal/methods , Humans
14.
J Biomed Mater Res A ; 67(3): 938-43, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14613242

ABSTRACT

Chitosan is considered to be a very promising biopolymer for various biomedical and pharmaceutical uses because of its nontoxic and biocompatible natures (Chandy T, Shama P. Biomater Artif Cells Artif Org 1990;18:1-24). In this study, we prepared porous chitosan scaffolds by lyophilization of chitosan solution. The scaffolds were modified with water-soluble polyanionic species such as alginate and heparin. The pore structures of these scaffolds were viewed via light and scanning electron microscopy. The scaffolds prepared have a high porosity of approximately 90% with mean pore sizes from 50 to 200 microm. They were used as substrates for hepatocytes culture. The cell attachment ratio was much higher than on monolayer membrane and hepatocytes exhibited a round cellular morphology with many microvilli evident on the surface of the cells. Metabolic activities of the cells were evaluated in terms of albumin secretion and urea synthesis. It was found that hepatocytes cultured on the modified scaffolds showed an increase in albumin secretion during the first 4 days and were more stable than those on monolayer membrane and nonmodified scaffolds. Therefore, primary rat hepatocytes cultured on modified scaffolds would be beneficial to liver assist device.


Subject(s)
Chitin/analogs & derivatives , Hepatocytes/cytology , Tissue Engineering , Animals , Biocompatible Materials , Cell Adhesion , Cell Culture Techniques/methods , Chitosan , Freeze Drying , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Liver, Artificial , Male , Porosity , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL