Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
FASEB J ; 29(4): 1208-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466901

ABSTRACT

We have previously shown that TNF-tumor necrosis factor receptor-2/p75 (TNFR2/p75) signaling plays a critical role in ischemia-induced neovascularization in skeletal muscle and heart tissues. To determine the role of TNF-TNFR2/p75 signaling in ischemia-induced inflammation and muscle regeneration, we subjected wild-type (WT) and TNFR2/p75 knockout (p75KO) mice to hind limb ischemia (HLI) surgery. Ischemia induced significant and long-lasting inflammation associated with considerable decrease in satellite-cell activation in p75KO muscle tissue up to 10 d after HLI surgery. To determine the possible additive negative roles of tissue aging and the absence of TNFR2/p75, either in the tissue or in the bone marrow (BM), we generated 2 chimeric BM transplantation (BMT) models where both young green fluorescent protein (GFP)-positive p75KO and WT BM-derived cells were transplanted into adult p75KO mice. HLI surgery was performed 1 mo after BMT, after confirming complete engraftment of the recipient BM with GFP donor cells. In adult p75KO with the WT-BMT, proliferative (Ki67(+)) cells were detected only by d 28 and were exclusively GFP(+), suggesting significantly delayed contribution of young WT-BM cell to adult p75KO ischemic tissue recovery. No GFP(+) young p75KO BM cells survived in adult p75KO tissue, signifying the additive negative roles of tissue aging combined with decreased/absent TNFR2/p75 signaling in postischemic recovery.


Subject(s)
Ischemia/pathology , Ischemia/physiopathology , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/physiology , Animals , Apoptosis , Bone Marrow Transplantation , Cell Proliferation , Disease Models, Animal , Green Fluorescent Proteins/genetics , Hindlimb/blood supply , Inflammation/etiology , Inflammation/pathology , Inflammation/physiopathology , Ischemia/therapy , Macrophages/pathology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic , Neutrophil Infiltration , Receptors, Tumor Necrosis Factor, Type II/metabolism , Recombinant Proteins/genetics , Regeneration , Signal Transduction
2.
J Biol Chem ; 289(20): 14178-93, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24711449

ABSTRACT

TNF-α, a pro-inflammatory cytokine, is highly expressed after being irradiated (IR) and is implicated in mediating radiobiological bystander responses (RBRs). Little is known about specific TNF receptors in regulating TNF-induced RBR in bone marrow-derived endothelial progenitor cells (BM-EPCs). Full body γ-IR WT BM-EPCs showed a biphasic response: slow decay of p-H2AX foci during the initial 24 h and increase between 24 h and 7 days post-IR, indicating a significant RBR in BM-EPCs in vivo. Individual TNF receptor (TNFR) signaling in RBR was evaluated in BM-EPCs from WT, TNFR1/p55KO, and TNFR2/p75KO mice, in vitro. Compared with WT, early RBR (1-5 h) were inhibited in p55KO and p75KO EPCs, whereas delayed RBR (3-5 days) were amplified in p55KO EPCs, suggesting a possible role for TNFR2/p75 signaling in delayed RBR. Neutralizing TNF in γ-IR conditioned media (CM) of WT and p55KO BM-EPCs largely abolished RBR in both cell types. ELISA protein profiling of WT and p55KO EPC γ-IR-CM over 5 days showed significant increases in several pro-inflammatory cytokines, including TNF-α, IL-1α (Interleukin-1 alpha), RANTES (regulated on activation, normal T cell expressed and secreted), and MCP-1. In vitro treatments with murine recombinant (rm) TNF-α and rmIL-1α, but not rmMCP-1 or rmRANTES, increased the formation of p-H2AX foci in nonirradiated p55KO EPCs. We conclude that TNF-TNFR2 signaling may induce RBR in naïve BM-EPCs and that blocking TNF-TNFR2 signaling may prevent delayed RBR in BM-EPCs, conceivably, in bone marrow milieu in general.


Subject(s)
Bone Marrow Cells/cytology , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Bystander Effect/drug effects , Bystander Effect/radiation effects , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/radiation effects , Gene Knockout Techniques , Histones/metabolism , Insulin-Like Growth Factor I/metabolism , Interleukin-1alpha/pharmacology , Ligands , Mice , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
3.
Am J Physiol Heart Circ Physiol ; 309(11): H1947-63, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26408534

ABSTRACT

There are 160,000 cancer patients worldwide treated with particle radiotherapy (RT). With the advent of proton, and high (H) charge (Z) and energy (E) HZE ionizing particle RT, the cardiovascular diseases risk estimates are uncertain. In addition, future deep space exploratory-type missions will expose humans to unknown but low doses of particle irradiation (IR). We examined molecular responses using transcriptome profiling in left ventricular murine cardiomyocytes isolated from mice that were exposed to 90 cGy, 1 GeV proton ((1)H) and 15 cGy, 1 GeV/nucleon iron ((56)Fe) over 28 days after exposure. Unsupervised clustering analysis of gene expression segregated samples according to the IR response and time after exposure, with (56)Fe-IR showing the greatest level of gene modulation. (1)H-IR showed little differential transcript modulation. Network analysis categorized the major differentially expressed genes into cell cycle, oxidative responses, and transcriptional regulation functional groups. Transcriptional networks identified key nodes regulating expression. Validation of the signal transduction network by protein analysis and gel shift assay showed that particle IR clearly regulates a long-lived signaling mechanism for ERK1/2, p38 MAPK signaling and identified NFATc4, GATA4, STAT3, and NF-κB as regulators of the response at specific time points. These data suggest that the molecular responses and gene expression to (56)Fe-IR in cardiomyocytes are unique and long-lasting. Our study may have significant implications for the efforts of National Aeronautics and Space Administration to develop heart disease risk estimates for astronauts and for patients receiving conventional and particle RT via identification of specific HZE-IR molecular markers.


Subject(s)
Gene Regulatory Networks/radiation effects , Iron Radioisotopes/toxicity , Myocytes, Cardiac/radiation effects , Radiotherapy, High-Energy/adverse effects , Signal Transduction/radiation effects , Animals , Cells, Cultured , Cluster Analysis , Enzyme Activation , Fibrosis , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/radiation effects , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Risk Assessment , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Time Factors , Transcription, Genetic/radiation effects , Transcriptome/radiation effects , Whole-Body Irradiation
4.
Sci Rep ; 14(1): 14127, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898063

ABSTRACT

Since conventional PID (Proportional-Integral-Derivative) controllers hardly control the robot to stabilize for constant force grinding under changing environmental conditions, it is necessary to add a compensation term to conventional PID controllers. An optimal parameter finding algorithm based on SAC (Soft-Actor-Critic) is proposed to solve the problem that the compensation term parameters are difficult to obtain, including training state action and normalization preprocessing, reward function design, and targeted deep neural network design. The algorithm is used to find the optimal controller compensation term parameters and applied to the PID controller to complete the compensation through the inverse kinematics of the robot to achieve constant force grinding control. To verify the algorithm's feasibility, a simulation model of a grinding robot with sensible force information is established, and the simulation results show that the controller trained with the algorithm can achieve constant force grinding of the robot. Finally, the robot constant force grinding experimental system platform is built for testing, which verifies the control effect of the optimal parameter finding algorithm on the robot constant force grinding and has specific environmental adaptability.

5.
Mol Ther Methods Clin Dev ; 32(1): 101191, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38352271

ABSTRACT

Despite the implementation of lifesaving newborn screening programs and a galactose-restricted diet, many patients with classic galactosemia develop long-term debilitating neurological deficits and primary ovarian insufficiency. Previously, we showed that the administration of human GALT mRNA predominantly expressed in the GalT gene-trapped mouse liver augmented the expression of hepatic GALT activity, which decreased not only galactose-1 phosphate (gal-1P) in the liver but also peripheral tissues. Since each peripheral tissue requires distinct methods to examine the biomarker and/or GALT effect, this highlights the necessity for alternative strategies to evaluate the overall impact of therapies. In this study, we established that whole-body galactose oxidation (WBGO) as a robust, noninvasive, and specific method to assess the in vivo pharmacokinetic and pharmacodynamic parameters of two experimental gene-based therapies that aimed to restore GALT activity in a mouse model of galactosemia. Although our results illustrated the long-lasting efficacy of AAVrh10-mediated GALT gene transfer, we found that GALT mRNA therapy that targets the liver predominantly is sufficient to sustain WBGO. The latter could have important implications in the design of novel targeted therapy to ensure optimal efficacy and safety.

6.
J Phys Chem Lett ; 14(49): 10920-10929, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38033191

ABSTRACT

Understanding and controlling carrier dynamics in two-dimensional (2D) van der Waals heterostructures through strain are crucial for their flexible applications. Here, femtosecond transient absorption spectroscopy is employed to elucidate the interlayer electron transfer and relaxation dynamics under external tensile strains in a WSe2/MoS2 heterostructure. The results show that a modest ∼1% tensile strain can significantly alter the lifetimes of electron transfer and nonradiative electron-hole recombination by >30%. Ab initio non-adiabatic molecular dynamics simulations suggest that tensile strain weakens the electron-phonon coupling, thereby suppressing the transfer and recombination dynamics. Theoretical predictions indicate that strain-induced energy difference increases along the electron transfer path could contribute to the prolongation of the transfer lifetime. A subpicosecond decay process, related to hot-electron cooling, remains almost unaffected by strain. This study demonstrates the potential of tuning interlayer carrier dynamics through external strains, offering insights into flexible optoelectronic device design with 2D materials.

7.
Front Med (Lausanne) ; 9: 799083, 2022.
Article in English | MEDLINE | ID: mdl-35814753

ABSTRACT

Background: Uveal melanoma (UM) is the most frequent primary intraocular tumor in adults. This study aims to develop a nomogram and an individualized web-based calculator to predict the overall survival (OS) of elderly patients with UM. Methods: Patients aged more than 60 years and diagnosed with UM were derived from the Surveillance, Epidemiology, and End Results (SEER) database during 2004-2015. The selected patients were randomly divided into training and validation cohorts. In the training cohort, the univariate and multivariate Cox analyses were carried out to determine the independent prognostic factors, and the predictors were integrated to establish a nomogram for predicting the 1-, 2-, and 3-year OS of elderly patients with UM. The discrimination of the nomogram was validated by receiver operating characteristic (ROC) curves and the area under the curve (AUC). The clinical practicability and accuracy of the nomogram were evaluated by the calibration curves and decision curve analysis (DCA). A web-based survival calculator was then constructed using a fitted survival prediction model (https://yuexinupup.shinyapps.io/DynNomapp/). Results: A total of 1,427 patients with UM were included in this study. Age, T stage, N stage, M stage, marital status, sex, and radiotherapy (RT) were identified as independent prognostic factors. Based on the abovementioned factors, the nomogram was then constructed. The AUC values of the nomogram predicting 1-, 2-, and 3-year OS were 0.841, 0.801, and 0.768 in the training cohort, and 0.745, 0.717, and 0.710 in the validation cohort, respectively. The calibration curves and DCA also indicated the good performance of the predictive model. Conclusion: This study established and validated a novel nomogram risk stratification model and a web-based survival rate calculator that can dynamically predict the long-term OS for elderly patients with UM.

8.
Am J Physiol Cell Physiol ; 301(1): C21-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21451102

ABSTRACT

Identification of factors that direct embryonic stem (ES) cell (ESC) differentiation into functional cardiomyocytes is essential for successful use of ESC-based therapy for cardiac repair. Neuregulin-1 (NRG1) and microRNA play important roles in the cardiac differentiation of ESCs. Understanding how NRG1 regulates microRNA will provide new mechanistic insights into the role of NRG1 on ESCs. It may also lead to the discovery of novel microRNAs that are important for ESC cardiac differentiation. The objective of this study was to assess the microRNA expression profile during NRG1-induced ESC cardiac differentiation. Murine ESCs were incubated with a recombinant NRG1ß or an inhibitor of ErbB2 or ErbB4 during hanging drop-induced cardiac differentiation. The expression of cardiac-specific markers and microRNAs was analyzed by RT-PCR and microRNA array, respectively. We found that the expression of NRG1 and the ErbB receptors was increased during hanging drop-induced cardiac differentiation of ESCs. NRG1 stimulation during a specific developmental window enhanced, while inhibition of the ErbB2 or ErbB4 receptor inhibited, cardiac differentiation of ESCs. NRG1 increased the expression of mmu-miR-296-3p and mmu-miR-200c*, and decreased mmu-miR-465b-5p. Inhibition of mmu-miR-296-3p or mmu-miR-200c* decreased, while inhibition of mmu-miR-465-5p increased, the differentiation of ESCs into the cardiac lineage. This is the first report demonstrating that microRNAs are differentially regulated by NRG1-ErbB signaling during cardiac differentiation of ESCs. This study has also identified new microRNAs that are important for ESC cardiac differentiation.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neuregulin-1/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Heart/growth & development , Mesoderm/embryology , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Microarray Analysis , Neuregulin-1/genetics , Oncogene Proteins v-erbB/antagonists & inhibitors , Oncogene Proteins v-erbB/metabolism , Polymerase Chain Reaction , Regeneration , Signal Transduction
10.
J Biomed Biotechnol ; 2011: 141039, 2011.
Article in English | MEDLINE | ID: mdl-21541238

ABSTRACT

The actions of Angiotensin II have been implicated in many cardiovascular conditions. It is widely accepted that the cardiovascular effects of Angiotensin II are mediated by different subtypes of receptors: AT(1) and AT(2). These membrane-bound receptors share a part of their nucleic acid but seem to have different distribution and pathophysiological actions. AT(1) mediates most of the Angiotensin II actions since it is ubiquitously expressed in the cardiovascular system of the normal adult. Moreover AT(2) is highly expressed in the developing fetus but its expression in the cardiovascular system is low and declines after birth. However the expression of AT(2) appears to be modulated by pathological states such as hypertension, myocardial infarction or any pathology associated to tissue remodeling or inflammation. The specific role of this receptor is still unclear and different studies involving in vivo and in vitro experiments have shown conflicting data. It is essential to clarify the role of the AT(2) receptor in the different pathological states as it is a potential site for an effective therapeutic regimen that targets the Angiotensin II system. We will review the different genetically modified mouse models used to study the AT(2) receptor and its association with cardiac hypertrophy and heart failure.


Subject(s)
Cardiomegaly/complications , Cardiomegaly/metabolism , Disease Models, Animal , Heart Failure/complications , Heart Failure/metabolism , Receptor, Angiotensin, Type 2/metabolism , Animals , Mice , Mice, Transgenic
11.
Nat Commun ; 11(1): 5339, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087718

ABSTRACT

Propionic acidemia/aciduria (PA) is an ultra-rare, life-threatening, inherited metabolic disorder caused by deficiency of the mitochondrial enzyme, propionyl-CoA carboxylase (PCC) composed of six alpha (PCCA) and six beta (PCCB) subunits. We herein report an enzyme replacement approach to treat PA using a combination of two messenger RNAs (mRNAs) (dual mRNAs) encoding both human PCCA (hPCCA) and PCCB (hPCCB) encapsulated in biodegradable lipid nanoparticles (LNPs) to produce functional PCC enzyme in liver. In patient fibroblasts, dual mRNAs encoded proteins localize in mitochondria and produce higher PCC enzyme activity vs. single (PCCA or PCCB) mRNA alone. In a hypomorphic murine model of PA, dual mRNAs normalize ammonia similarly to carglumic acid, a drug approved in Europe for the treatment of hyperammonemia due to PA. Dual mRNAs additionally restore functional PCC enzyme in liver and thus reduce primary disease-associated toxins in a dose-dependent manner in long-term 3- and 6-month repeat-dose studies in PA mice. Dual mRNAs are well-tolerated in these studies with no adverse findings. These studies demonstrate the potential of mRNA technology to chronically administer multiple mRNAs to produce large complex enzymes, with applicability to other genetic disorders.


Subject(s)
Enzyme Replacement Therapy/methods , Propionic Acidemia/therapy , RNA, Messenger/therapeutic use , Animals , Disease Models, Animal , Glutamates/therapeutic use , Humans , Kinetics , Lipids/chemistry , Liver/enzymology , Methylmalonyl-CoA Decarboxylase/chemistry , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/enzymology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Propionic Acidemia/genetics , Propionic Acidemia/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics
12.
Am J Physiol Heart Circ Physiol ; 297(6): H1974-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19801490

ABSTRACT

Neuregulin-1 (NRG1) is a potential therapeutic agent for the treatment of doxorubicin (Dox)-induced heart failure. NRG1, however, activates the erbB2 receptor, which is frequently overexpressed in breast cancers. It is, therefore, important to understand how NRG1, via erbB2, protects the heart against Dox cardiotoxicity. Here, we studied NRG1-erbB2 signaling in Dox-treated mice hearts and in isolated neonatal rat ventricular myocytes (NRVM). Male C57BL/6 mice were treated with recombinant NRG1 before and daily after a single dose of Dox. Cardiac function was determined by catheterization. Two-week survival was analyzed by the Kaplan-Meier method. Cardiac troponins [cardiac troponin I (cTnI) and cardiac troponin T (cTnT)] and phosphorylated Akt protein levels were determined in mice hearts and in NRVM by Western blot analysis. Activation of caspases and ubiquitinylation of troponins were determined in NRVM by caspase assay and immunoprecipitation. NRG1 significantly improved survival and cardiac function in Dox-treated mice. NRG1 reduced the decrease in cTnI, cTnT, and cardiac troponin C (cTnC) and maintained Akt phosphorylation in Dox-treated mice hearts. NRG1 reduced the decrease in cTnI and cTnT mRNA and proteins in Dox-treated NRVM. Inhibition of erbB2, phosphoinositide 3-kinase (PI3K), Akt, and mTOR blocked the protective effects of NRG1 on cTnI and cTnT in NRVM. NRG1 significantly reduced Dox-induced caspase activation, which degraded troponins, in NRVM. NRG1 reduced Dox-induced proteasome degradation of cTnI. NRG1 attenuates Dox-induced decrease in cardiac troponins by increasing transcription and translation and by inhibiting caspase activation and proteasome degradation of troponin proteins. NRG1 maintains cardiac troponins by the erbB2-PI3K pathway, which may lessen Dox-induced cardiac dysfunction.


Subject(s)
Cardiovascular Agents/administration & dosage , Heart Diseases/prevention & control , Myocardium/metabolism , Neuregulin-1/administration & dosage , Signal Transduction/drug effects , Troponin I/metabolism , Troponin T/metabolism , Animals , Animals, Newborn , Antibiotics, Antineoplastic , Biomarkers/blood , Caspases/metabolism , Cells, Cultured , Creatine Kinase/blood , Disease Models, Animal , Doxorubicin , Glycoproteins/metabolism , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/physiopathology , Humans , Injections, Subcutaneous , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptor, ErbB-2 , Recombinant Proteins/administration & dosage , TOR Serine-Threonine Kinases , Time Factors , Transcription, Genetic/drug effects , Troponin I/blood , Troponin I/genetics , Troponin T/blood , Troponin T/genetics , Ubiquitination , Up-Regulation , Ventricular Function, Left/drug effects
13.
J Thorac Dis ; 11(11): 4551-4561, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31903244

ABSTRACT

BACKGROUND: To investigate the relationship between pulmonary function and brachial-ankle pulse wave velocity (baPWV). METHODS: A cross-sectional study was conducted. A total of 11,388 people with complete pulmonary function test and baPWV data and who participated in both the health examination of the Kailuan Occupational Disease Prevention and Treatment Center in 2014-2016 and the health checkup of the Kailuan Group in 2012 and 2014 were selected as subjects. The study population was divided into four groups by forced vital capacity (FVC) quartiles (group 1: FVC <3.50 L; group 2: 3.50 L ≤ FVC <3.96 L; group 3: 3.96 L ≤ FVC <4.47 L; group 4: FVC ≥4.47 L) and divided into four groups by forced expiratory volume in one second (FEV1) quartile (group 1: FEV1 <3.15 L; group 2: 3.15 L ≤ FVC <3.61 L; group 3: 3.61 L ≤ FVC <4.08 L; group 4: FVC ≥4.08 L). Linear regression analysis and multivariate logistic regression were used to analyze the effects of pulmonary function on baPWV. RESULTS: When grouped by FVC, the baPWV of the first group was significantly higher than the other groups. Similarly, the incidence of arteriosclerosis in the first group was significantly higher than the other groups. When grouped by FEV1, the baPWV of the first group was significantly higher than the other groups. The incidence of arteriosclerosis was also significantly higher in the first group than the other groups. After correcting for other confounding factors using linear regression, it was found that the effects of FVC and FEV1 on the study subject's baPWV were -23.84 and -24.65 L, respectively. Multivariate logistic regression analysis showed that when grouped by FVC quartile, the risk of arteriosclerosis was increased by 34% in group 1 compared with group 4 (OR: 1.34, 95% CI: 1.17-1.52); the risk of arteriosclerosis was increased by 16% in group 2 compared with group 4 (OR: 1.16, 95% CI: 1.03-1.31). When grouped by the FEV1 quartile, the risk of arteriosclerosis was increased by 25% in group 1 compared with group 4 (OR: 1.25, 95% CI: 1.10-1.42). CONCLUSIONS: Decreased pulmonary function is negatively correlated with baPWV and is an independent risk factor for arteriosclerosis.

14.
Eur J Pharmacol ; 592(1-3): 123-7, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18627770

ABSTRACT

Clozapine, an atypical antipsychotic, is very effective in the treatment of resistant schizophrenia. However, cardiotoxicity of clozapine, particularly in young patients, has raised concerns about its safety. Increased catecholamines have been postulated to trigger an inflammatory response resulting in myocarditis, dilated cardiomyopathy, and death, although this has not yet been thoroughly studied. Here, we used the mouse to study whether clozapine administration could cause adverse myocarditis associated with an increase in catecholamines. Male Balb/C mice, age ~6 weeks, were administered 5, 10 or 25 mg/kg clozapine daily for 7 and 14 days; one group was administered 25 mg/kg clozapine plus 2 mg/kg propranolol for 14 days. Saline-treated mice served as controls. Heart sections were stained with hematoxylin and eosin for histopathological examination. Plasma catecholamines were measured with HPLC. Myocardial TNF-alpha concentrations were determined by ELISA. Histopathology of clozapine-treated mice showed a significant dose-related increase in myocardial inflammation that correlated with plasma catecholamine levels and release of TNF-alpha. Propranolol significantly attenuated these effects. A hypercatecholaminergic state induced by clozapine could explain the occurrence of myocarditis in some patients. Our data suggest that a beta-adrenergic blocking agent may be effective in reducing the incidence and severity of clozapine-induced myocarditis.


Subject(s)
Antipsychotic Agents/toxicity , Catecholamines/physiology , Clozapine/toxicity , Myocarditis/chemically induced , Myocarditis/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred BALB C , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Propranolol/pharmacology , Tumor Necrosis Factor-alpha/metabolism
15.
Radiat Res ; 188(2): 191-203, 2017 08.
Article in English | MEDLINE | ID: mdl-28613990

ABSTRACT

Deep-space travel presents risks of exposure to ionizing radiation composed of a spectrum of low-fluence protons (1H) and high-charge and energy (HZE) iron nuclei (e.g., 56Fe). When exposed to galactic cosmic rays, each cell in the body may be traversed by 1H every 3-4 days and HZE nuclei every 3-4 months. The effects of low-dose sequential fractionated 1H or HZE on the heart are unknown. In this animal model of simulated ionizing radiation, middle-aged (8-9 months old) male C57BL/6NT mice were exposed to radiation as follows: group 1, nonirradiated controls; group 2, three fractionated doses of 17 cGy 1H every other day (1H × 3); group 3, three fractionated doses of 17 cGy 1H every other day followed by a single low dose of 15 cGy 56Fe two days after the final 1H dose (1H × 3 + 56Fe); and group 4, a single low dose of 15 cGy 56Fe followed (after 2 days) by three fractionated doses of 17 cGy 1H every other day (56Fe + 1H × 3). A subgroup of mice from each group underwent myocardial infarction (MI) surgery at 28 days postirradiation. Cardiac structure and function were assessed in all animals at days 7, 14 and 28 after MI surgery was performed. Compared to the control animals, the treatments that groups 2 and 3 received did not induce negative effects on cardiac function or structure. However, compared to all other groups, the animals in group 4, showed depressed left ventricular (LV) functions at 1 month with concomitant enhancement in cardiac fibrosis and induction of cardiac hypertrophy signaling at 3 months. In the irradiated and MI surgery groups compared to the control group, the treatments received by groups 2 and 4 did not induce negative effects at 1 month postirradiation and MI surgery. However, in group 3 after MI surgery, there was a 24% increase in mortality, significant decreases in LV function and a 35% increase in post-infarction size. These changes were associated with significant decreases in the angiogenic and cell survival signaling pathways. These data suggest that fractionated doses of radiation induces cellular and molecular changes that result in depressed heart functions both under basal conditions and particularly after myocardial infarction.


Subject(s)
Dose Fractionation, Radiation , Heart/physiology , Heart/radiation effects , Iron/adverse effects , Protons/adverse effects , Animals , Cosmic Radiation/adverse effects , Dose-Response Relationship, Radiation , Heart/physiopathology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology
17.
Circulation ; 107(8): 1176-82, 2003 Mar 04.
Article in English | MEDLINE | ID: mdl-12615798

ABSTRACT

BACKGROUND: Chronic cardiac unloading of the normal heart results in the reduction of left ventricular (LV) mass, but effects on myocyte contractile function are not known. METHODS AND RESULTS: Cardiac unloading and reduction in LV mass were induced by heterotopic heart transplantation to the abdominal aorta in isogenic rats. Contractility and [Ca(2+)](i) regulation in LV myocytes were studied at both 2 and 5 weeks after transplantation. Native in situ hearts from recipient animals were used as the controls for all experiments. Contractile function indices in myocytes from 2-week unloaded and native (control) hearts were similar under baseline conditions (0.5 Hz, 1.2 mmol/L [Ca(2+)](o), and 36 degrees C) and in response to stimulation with high [Ca(2+)](o) (range 2.5 to 4.0 mmol/L). In myocytes from 5-week unloaded hearts, there were no differences in fractional cell shortening and peak-systolic [Ca(2+)](i) at baseline; however, time to 50% relengthening and time to 50% decline in [Ca(2+)](i) were prolonged compared with controls. Severe defects in fractional cell shortening and peak-systolic [Ca(2+)](i) were elicited in myocytes from 5-week unloaded hearts in response to high [Ca(2+)](o). However, there were no differences in the contractile response to isoproterenol between myocytes from unloaded and native hearts. In 5-week unloaded hearts, but not in 2-week unloaded hearts, LV protein levels of phospholamban were increased (345% of native heart values). Protein levels of sarcoplasmic reticulum Ca(2+) ATPase and the Na(+)/Ca(2+) exchanger were not changed. CONCLUSIONS: Chronic unloading of the normal heart caused a time-dependent depression of myocyte contractile function, suggesting the potential for impaired performance in states associated with prolonged cardiac atrophy.


Subject(s)
Calcium/analysis , Myocardial Contraction , Myocytes, Cardiac/physiology , Animals , Cardiomegaly/metabolism , Cells, Cultured , Heart Transplantation , Heart Ventricles/metabolism , Kinetics , Male , Muscle Proteins/metabolism , Muscular Atrophy/physiopathology , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/cytology , Rats , Rats, Inbred Lew , Ventricular Remodeling
18.
Circulation ; 110(6): 713-7, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15289373

ABSTRACT

BACKGROUND: Neuregulins are required for maintenance of acetylcholine receptor-inducing activity of nicotinic receptors in neurons and skeletal muscle, but effects of neuregulins on muscarinic receptors are not known. In the normal heart, parasympathetic activation counterbalances beta-adrenergic activation. To test the hypothesis that neuregulins modify parasympathetic function in the heart, we studied cardiomyocytes from mice heterozygous for neuregulin-1 gene deletion (NRG-1+/-) and examined the effects of beta-adrenergic stimulation on contractility in the presence and absence of the muscarinic agonist carbachol. METHODS AND RESULTS: We evaluated contraction and intracellular Ca2+ transients ([Ca2+]i) in left ventricular (LV) myocytes loaded with Fluo-3 from NRG-1+/- and wild-type (WT) mice. Under baseline conditions (0.5 Hz, 1.5 mmol/L [Ca2+]o, 25 degrees C), characteristics of myocyte contraction/relengthening and systolic/diastolic [Ca2+]i were not different between WT and NRG-1+/- mice. The steady-state increases in fractional shortening (FS) and peak-systolic [Ca2+]i in response to isoproterenol were similar in both groups. In WT myocytes stimulated with isoproterenol, carbachol decreased FS, peak-systolic [Ca2+]i, and cAMP levels. In NRG-1+/- myocytes, carbachol did not attenuate either FS or peak-systolic [Ca2+]i, associated with the failure to decrease cAMP levels. Investigation of muscarinic receptor signaling showed no difference of LV protein levels of muscarinic M2 receptors or G protein Galpha(i1,2), Galpha(i3), and Galpha(o) subunits. CONCLUSIONS: Cardiomyocytes deficient in neuregulin signaling are unable to adequately counterbalance beta-adrenergic activation by inhibitory parasympathetic activity. This mechanism may contribute to the known increased risk of heart failure in injured human hearts when neuregulin signaling is suppressed.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Neuregulin-1/physiology , Receptor, Muscarinic M2/physiology , Animals , Calcium Signaling/drug effects , Carbachol/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/physiology , Cyclic AMP/physiology , Gene Deletion , Heart Conduction System/drug effects , Heart Conduction System/physiology , Heterozygote , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Muscarinic Agonists/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Neuregulin-1/deficiency , Neuregulin-1/genetics , Parasympathetic Nervous System/drug effects , Parasympathetic Nervous System/physiology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/physiology
19.
Stem Cells Int ; 2015: 496512, 2015.
Article in English | MEDLINE | ID: mdl-26074973

ABSTRACT

Bone-marrow- (BM-) derived endothelial progenitor cells (EPCs) are critical for endothelial cell maintenance and repair. During future space exploration missions astronauts will be exposed to space irradiation (IR) composed of a spectrum of low-fluence protons ((1)H) and high charge and energy (HZE) nuclei (e.g., iron-(56)Fe) for extended time. How the space-type IR affects BM-EPCs is limited. In media transfer experiments in vitro we studied nontargeted effects induced by (1)H- and (56)Fe-IR conditioned medium (CM), which showed significant increase in the number of p-H2AX foci in nonirradiated EPCs between 2 and 24 h. A 2-15-fold increase in the levels of various cytokines and chemokines was observed in both types of IR-CM at 24 h. Ex vivo analysis of BM-EPCs from single, low-dose, full-body (1)H- and (56)Fe-IR mice demonstrated a cyclical (early 5-24 h and delayed 28 days) increase in apoptosis. This early increase in BM-EPC apoptosis may be the effect of direct IR exposure, whereas late increase in apoptosis could be a result of nontargeted effects (NTE) in the cells that were not traversed by IR directly. Identifying the role of specific cytokines responsible for IR-induced NTE and inhibiting such NTE may prevent long-term and cyclical loss of stem and progenitors cells in the BM milieu.

20.
Front Oncol ; 5: 231, 2015.
Article in English | MEDLINE | ID: mdl-26528440

ABSTRACT

Exposure of individuals to ionizing radiation (IR), as in the case of astronauts exploring space or radiotherapy cancer patients, increases their risk of developing secondary cancers and other health-related problems. Bone marrow (BM), the site in the body where hematopoietic stem cell (HSC) self-renewal and differentiation to mature blood cells occurs, is extremely sensitive to low-dose IR, including irradiation by high-charge and high-energy particles. Low-dose IR induces DNA damage and persistent oxidative stress in the BM hematopoietic cells. Inefficient DNA repair processes in HSC and early hematopoietic progenitors can lead to an accumulation of mutations whereas long-lasting oxidative stress can impair hematopoiesis itself, thereby causing long-term damage to hematopoietic cells in the BM niche. We report here that low-dose (1)H- and (56)Fe-IR significantly decreased the hematopoietic early and late multipotent progenitor (E- and L-MPP, respectively) cell numbers in mouse BM over a period of up to 10 months after exposure. Both (1)H- and (56)Fe-IR increased the expression of pluripotent stem cell markers Sox2, Nanog, and Oct4 in L-MPPs and 10 months post-IR exposure. We postulate that low doses of (1)H- and (56)Fe-IR may induce endogenous cellular reprogramming of BM hematopoietic progenitor cells to assume a more primitive pluripotent phenotype and that IR-induced oxidative DNA damage may lead to mutations in these BM progenitors. This could then be propagated to successive cell lineages. Persistent impairment of BM progenitor cell populations can disrupt hematopoietic homeostasis and lead to hematologic disorders, and these findings warrant further mechanistic studies into the effects of low-dose IR on the functional capacity of BM-derived hematopoietic cells including their self-renewal and pluripotency.

SELECTION OF CITATIONS
SEARCH DETAIL